Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 120
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 109(1): 116-135, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34965383

ABSTRACT

The high-grade serous ovarian cancer (HGSOC) risk locus at chromosome 1p34.3 resides within a frequently amplified genomic region signifying the presence of an oncogene. Here, we integrate in silico variant-to-function analysis with functional studies to characterize the oncogenic potential of candidate genes in the 1p34.3 locus. Fine mapping of genome-wide association statistics identified candidate causal SNPs local to H3K27ac-demarcated enhancer regions that exhibit allele-specific binding for CTCF in HGSOC and normal fallopian tube secretory epithelium cells (FTSECs). SNP risk associations colocalized with eQTL for six genes (DNALI1, GNL2, RSPO1, SNIP1, MEAF6, and LINC01137) that are more highly expressed in carriers of the risk allele, and three (DNALI1, GNL2, and RSPO1) were upregulated in HGSOC compared to normal ovarian surface epithelium cells and/or FTSECs. Increased expression of GNL2 and MEAF6 was associated with shorter survival in HGSOC with 1p34.3 amplifications. Despite its activation of Ɵ-catenin signaling, RSPO1 overexpression exerted no effects on proliferation or colony formation in our study of ovarian cancer and FTSECs. Instead, GNL2, MEAF6, and SNIP1 silencing impaired inĀ vitro ovarian cancer cell growth. Additionally, GNL2 silencing diminished xenograft tumor formation, whereas overexpression stimulated proliferation and colony formation in FTSECs. GNL2 influences 60S ribosomal subunit maturation and global protein synthesis in ovarian cancer and FTSECs, providing a potential mechanism of how GNL2 upregulation might promote ovarian cancer development and mediate genetic susceptibility of HGSOC.


Subject(s)
Chromosomes, Human, Pair 1 , Cystadenocarcinoma, Serous/genetics , GTP-Binding Proteins/genetics , Genetic Predisposition to Disease , Ovarian Neoplasms/genetics , Quantitative Trait Loci , Alleles , Alternative Splicing , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromatin Immunoprecipitation Sequencing , Cystadenocarcinoma, Serous/pathology , DNA Copy Number Variations , Disease Models, Animal , Enhancer Elements, Genetic , Female , GTP-Binding Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Genetic Association Studies , Genome-Wide Association Study , Heterografts , Humans , Mice , Neoplasm Grading , Odds Ratio , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Polymorphism, Single Nucleotide , Prognosis , Transcriptome , White People
2.
Lab Invest ; 104(5): 102041, 2024 May.
Article in English | MEDLINE | ID: mdl-38431116

ABSTRACT

A specific splicing isoform of RNASET2 is associated with worse oncologic outcomes in clear cell renal cell carcinoma (ccRCC). However, the interplay between wild-type RNASET2 and its splice variant and how this might contribute to the pathogenesis of ccRCC remains poorly understood. We sought to better understand the relationship of RNASET2 in the pathogenesis of ccRCC and the interplay with a pathogenic splicing isoform (RNASET2-SV) and the tumor immune microenvironment. Using data from The Cancer Genome Atlas and Clinical Proteomic Tumor Analysis Consortium, we correlated clinical variables to RNASET2 expression and the presence of a specific RNASET2-SV. Immunohistochemical staining with matched RNA sequencing of ccRCC patients was then utilized to understand the spatial relationships of RNASET2 with immune cells. Finally, inĀ vitro studies were performed to demonstrate the oncogenic role of RNASET2 and highlight its potential mechanisms. RNASET2 gene expression is associated with higher grade tumors and worse overall survival in The Cancer Genome Atlas cohort. The presence of the RNASET2-SV was associated with increased expression of the wild-type RNASET2 protein and epigenetic modifications of the gene. Immunohistochemical staining revealed increased intracellular accumulation of RNASET2 in patients with increased RNA expression of RNASET2-SV. InĀ vitro experiments reveal that this accumulation results in increased cell proliferation, potentially from altered metabolic pathways. RNASET2 exhibits a tumor-promoting role in the pathogenesis of ccRCC that is increased in the presence of a specific RNASET2-SV and associated with changes in the cellular localization of the protein.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Ribonucleases , Female , Humans , Male , Middle Aged , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Ribonucleases/genetics , Ribonucleases/metabolism , Tumor Microenvironment , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
BMC Bioinformatics ; 24(1): 164, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37095442

ABSTRACT

BACKGROUND: Massively parallel sequencing includes many liquid handling steps which introduce the possibility of sample swaps, mixing, and duplication. The unique profile of inherited variants in human genomes allows for comparison of sample identity using sequence data. A comparison of all samples vs. each other (all vs. all) provides both identification of mismatched samples and the possibility of resolving swapped samples. However, all vs. all comparison complexity grows as the square of the number of samples, so efficiency becomes essential. RESULTS: We have developed a tool for fast all vs. all genotype comparison using low level bitwise operations built into the Perl programming language. Importantly, we have also developed a complete workflow allowing users to start with either raw FASTQ sequence files, aligned BAM files, or genotype VCF files and automatically generate comparison metrics and summary plots. The tool is freely available at https://github.com/teerjk/TimeAttackGenComp/ . CONCLUSIONS: A fast and easy to use method for genotype comparison as described here is an important tool to ensure high quality and robust results in sequencing studies.


Subject(s)
High-Throughput Nucleotide Sequencing , Software , Humans , Workflow , Genotype , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods , DNA , Sequence Analysis, DNA/methods
4.
PLoS Genet ; 16(4): e1008642, 2020 04.
Article in English | MEDLINE | ID: mdl-32310940

ABSTRACT

Undifferentiated embryonal sarcoma of the liver (UESL) is a rare and aggressive malignancy. Though the molecular underpinnings of this cancer have been largely unexplored, recurrent chromosomal breakpoints affecting a noncoding region on chr19q13, which includes the chromosome 19 microRNA cluster (C19MC), have been reported in several cases. We performed comprehensive molecular profiling on samples from 14 patients diagnosed with UESL. Congruent with prior reports, we identified structural variants in chr19q13 in 10 of 13 evaluable tumors. From whole transcriptome sequencing, we observed striking expressional activity of the entire C19MC region. Concordantly, in 7 of 7 samples undergoing miRNAseq, we observed hyperexpression of the miRNAs within this cluster to levels >100 fold compared to matched normal tissue or a non-C19MC amplified cancer cell line. Concurrent TP53 mutation or copy number loss was identified in all evaluable tumors with evidence of C19MC overexpression. We find that C19MC miRNAs exhibit significant negative correlation to TP53 regulatory miRNAs and K-Ras regulatory miRNAs. Using RNA-seq we identified that pathways relevant to cellular differentiation as well as mRNA translation machinery are transcriptionally enriched in UESL. In summary, utilizing a combination of next-generation sequencing and high-density arrays we identify the combination of C19MC hyperexpression via chromosomal structural event with TP53 mutation or loss as highly recurrent genomic features of UESL.


Subject(s)
Chromosome Breakpoints , Chromosomes, Human, Pair 19/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Mutation , Neoplasms, Germ Cell and Embryonal/genetics , Sarcoma/genetics , Tumor Suppressor Protein p53/genetics , Aneuploidy , Child , Child, Preschool , Female , Genes, ras/genetics , Genomic Instability/genetics , Humans , Infant , Male , Transcription Initiation Site , Tumor Suppressor Protein p53/deficiency , Up-Regulation
5.
Semin Cancer Biol ; 61: 149-157, 2020 04.
Article in English | MEDLINE | ID: mdl-31689494

ABSTRACT

Acral melanomas arise on the non-hair bearing skin of the palms, soles and in the nail beds. These rare tumors comprise 2-3 % of all melanomas, are not linked to UV-exposure, and represent the most frequent subtype of melanomas in patients of Asian, African and Hispanic origin. Although recent work has revealed candidate molecular events that underlie acral melanoma development, this knowledge is not yet been translated into efficacious local, regional, or systemic therapies. In the current review, we describe the clinical characteristics of acral melanoma and outline the genetic basis of acral melanoma development. Further discussion is given to the current status of systemic therapy for acral melanoma with a focus on ongoing developments in both immunotherapy and targeted therapy for the treatment of advanced disease.


Subject(s)
Genetic Predisposition to Disease , Genomics , Melanoma/diagnosis , Melanoma/etiology , Melanoma/therapy , Skin Neoplasms/diagnosis , Skin Neoplasms/etiology , Skin Neoplasms/therapy , Biomarkers, Tumor , Disease Management , Genomics/methods , Humans , Melanoma/metabolism , Mutation , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Translational Research, Biomedical
6.
Lab Invest ; 101(2): 204-217, 2021 02.
Article in English | MEDLINE | ID: mdl-33037322

ABSTRACT

Pancreatic cancer (PaCa) is the third leading cause of cancer-related deaths in the United States. There is an unmet need to develop strategies to detect PaCa at an early, operable stage and prevent its progression. Intraductal papillary mucinous neoplasms (IPMNs) are cystic PaCa precursors that comprise nearly 50% of pancreatic cysts detected incidentally via cross-sectional imaging. Since IPMNs can progress from low- and moderate-grade dysplasia to high-grade dysplasia and invasion, the study of these lesions offers a prime opportunity to develop early detection and prevention strategies. Organoids are an ideal preclinical platform to study IPMNs, and the objective of the current investigation was to establish a living biobank of patient-derived organoids (PDO) from IPMNs. IPMN tumors and adjacent normal pancreatic tissues were successfully harvested from 15 patients with IPMNs undergoing pancreatic surgical resection at Moffitt Cancer Center & Research Institute (Tampa, FL) between May of 2017 and March of 2019. Organoid cultures were also generated from cryopreserved tissues. Organoid count and size were determined over time by both Image-Pro Premier 3D Version 9.1 digital platform and Matlab application of a Circular Hough Transform algorithm, and histologic and genomic characterization of a subset of the organoids was performed using immunohistochemistry and targeted sequencing, respectively. The success rates for organoid generation from IPMN tumor and adjacent normal pancreatic tissues were 81% and 87%, respectively. IPMN organoids derived from different epithelial subtypes showed different morphologies in vitro, and organoids recapitulated histologic and genomic characteristics of the parental IPMN tumor. In summary, this preclinical model has the potential to provide new opportunities to unveil mechanisms of IPMN progression to invasion and to shed insight into novel biomarkers for early detection and targets for chemoprevention.


Subject(s)
Biological Specimen Banks , Organoids/pathology , Pancreas/pathology , Pancreatic Intraductal Neoplasms/pathology , Aged , Aged, 80 and over , Algorithms , Cell Culture Techniques , Female , Histocytochemistry , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Organoids/cytology , Pancreas/cytology , Tissue Culture Techniques
7.
J Neurooncol ; 155(3): 319-324, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34669147

ABSTRACT

BACKGROUND: Risk factors for meningioma include female gender, African American race, high body mass index (BMI), and exposure to ionizing radiation. Although genome-wide association studies (GWAS) have identified two nuclear genome risk loci for meningioma (rs12770228 and rs2686876), the relation between mitochondrial DNA (mtDNA) sequence variants and meningioma is unknown. METHODS: We examined the association of 42 common germline mtDNA variants (minor allele frequency ≥ 5%), haplogroups, and genes with meningioma in 1080 controls and 478 meningioma cases from a case-control study conducted at medical centers in the southeastern United States. Associations were examined separately for meningioma overall and by WHO grade (n = 409 grade I and n = 69 grade II/III). RESULTS: Overall, meningioma was significantly associated with being female (OR 2.85; 95% CI 2.21-3.69), self-reported African American race (OR 2.38, 95% CI 1.41-3.99), and being overweight (OR 1.48; 95% CI 1.11-1.97) or obese (OR 1.70; 95% CI 1.25-2.31). The variant m.16362T > C (rs62581341) in the mitochondrial control region was positively associated with grade II/III meningiomas (OR 2.33; 95% CI 1.14-4.77), but not grade I tumors (OR 0.99; 95% CI 0.64-1.53). Haplogroup L, a marker for African ancestry, was associated with meningioma overall (OR 2.92; 95% CI 1.01-8.44). However, after stratifying by self-reported race, this association was only apparent among the few self-reported Caucasians with this haplogroup (OR 6.35; 95% CI 1.56-25.9). No other mtDNA variant, haplogroup, or gene was associated with meningioma. CONCLUSION: Common mtDNA variants and major mtDNA haplogroups do not appear to have associations with the odds of developing meningioma.


Subject(s)
Meningeal Neoplasms , Meningioma , Case-Control Studies , DNA, Mitochondrial/genetics , Female , Genome-Wide Association Study , Haplotypes , Humans , Meningeal Neoplasms/genetics , Meningioma/genetics , Polymorphism, Single Nucleotide
8.
Genes Chromosomes Cancer ; 57(1): 19-27, 2018 01.
Article in English | MEDLINE | ID: mdl-28891274

ABSTRACT

NF1 mutations predispose to neurofibromatosis type 1 (NF1) and women with NF1 have a moderately elevated risk for breast cancer, especially under age 50. Germline genomic analysis may better define the risk so screening and prevention can be applied to the individuals who benefit the most. Survey conducted in several neurofibromatosis clinics in the United States has demonstrated a 17.2% lifetime risk of breast cancer in women affected with NF1. Cumulated risk to age 50 is estimated to be 9.27%. For genomic profiling, fourteen women with NF1 and a history of breast cancer were recruited and underwent whole exome sequencing (WES), targeted genomic DNA based and RNA-based analysis of the NF1 gene. Deleterious NF1 pathogenic variants were identified in each woman. Frameshift mutations because of deletion/duplication/complex rearrangement were found in 50% (7/14) of the cases, nonsense mutations in 21% (3/14), in-frame splice mutations in 21% (3/14), and one case of missense mutation (7%, 1/14). No deleterious mutation was found in the following high/moderate-penetrance breast cancer genes: ATM, BRCA1, BRCA2, BARD1, BRIP1, CDH1, CHEK2, FANCC, MRE11A, NBN, PALB2, PTEN, RAD50, RAD51C, TP53, and STK11. Twenty-five rare or common variants in cancer related genes were discovered and may have contributed to the breast cancers in these individuals. Breast cancer predisposition modifiers in women with NF1 may involve a great variety of molecular and cellular functions.


Subject(s)
Breast Neoplasms/genetics , Exome Sequencing , Germ-Line Mutation , Neurofibromatosis 1/genetics , Adult , Breast Neoplasms/epidemiology , Female , Genes, Neurofibromatosis 1 , Humans , Middle Aged , Neurofibromatosis 1/complications , Oncogenes , Penetrance , Polymorphism, Single Nucleotide
9.
Hum Genomics ; 11(1): 22, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28870239

ABSTRACT

BACKGROUND: Observations of recurrent somatic mutations in tumors have led to identification and definition of signaling and other pathways that are important for cancer progression and therapeutic targeting. As tumor cells contain both an individual's inherited genetic variants and somatic mutations, challenges arise in distinguishing these events in massively parallel sequencing datasets. Typically, both a tumor sample and a "normal" sample from the same individual are sequenced and compared; variants observed only in the tumor are considered to be somatic mutations. However, this approach requires two samples for each individual. RESULTS: We evaluate a method of detecting somatic mutations in tumor samples for which only a subset of normal samples are available. We describe tuning of the method for detection of mutations in tumors, filtering to remove inherited variants, and comparison of detected mutations to several matched tumor/normal analysis methods. Filtering steps include the use of population variation datasets to remove inherited variants as well a subset of normal samples to remove technical artifacts. We then directly compare mutation detection with tumor-only and tumor-normal approaches using the same sets of samples. Comparisons are performed using an internal targeted gene sequencing dataset (nĀ =Ā 3380) as well as whole exome sequencing data from The Cancer Genome Atlas project (nĀ =Ā 250). Tumor-only mutation detection shows similar recall (43-60%) but lesser precision (20-21%) to current matched tumor/normal approaches (recall 43-73%, precision 30-82%) when compared to a "gold-standard" tumor/normal approach. The inclusion of a small pool of normal samples improves precision, although many variants are still uniquely detected in the tumor-only analysis. CONCLUSIONS: A detailed method for somatic mutation detection without matched normal samples enables study of larger numbers of tumor samples, as well as tumor samples for which a matched normal is not available. As sensitivity/recall is similar to tumor/normal mutation detection but precision is lower, tumor-only detection is more appropriate for classification of samples based on known mutations. Although matched tumor-normal analysis is preferred due to higher precision, we demonstrate that mutation detection without matched normal samples is possible for certain applications.


Subject(s)
DNA Mutational Analysis/methods , Neoplasms/genetics , Software , Databases, Factual , Genotype , High-Throughput Nucleotide Sequencing/methods , Humans , Mutation , Sensitivity and Specificity
10.
Brain ; 140(2): 370-386, 2017 02.
Article in English | MEDLINE | ID: mdl-28007986

ABSTRACT

Leukoencephalopathies are a group of white matter disorders related to abnormal formation, maintenance, and turnover of myelin in the central nervous system. These disorders of the brain are categorized according to neuroradiological and pathophysiological criteria. Herein, we have identified a unique form of leukoencephalopathy in seven patients presenting at ages 2 to 4 months with progressive microcephaly, spastic quadriparesis, and global developmental delay. Clinical, metabolic, and imaging characterization of seven patients followed by homozygosity mapping and linkage analysis were performed. Next generation sequencing, bioinformatics, and segregation analyses followed, to determine a loss of function sequence variation in the phospholipase A2-activating protein encoding gene (PLAA). Expression and functional studies of the encoded protein were performed and included measurement of prostaglandin E2 and cytosolic phospholipase A2 activity in membrane fractions of fibroblasts derived from patients and healthy controls. Plaa-null mice were generated and prostaglandin E2 levels were measured in different tissues. The novel phenotype of our patients segregated with a homozygous loss-of-function sequence variant, causing the substitution of leucine at position 752 to phenylalanine, in PLAA, which causes disruption of the protein's ability to induce prostaglandin E2 and cytosolic phospholipase A2 synthesis in patients' fibroblasts. Plaa-null mice were perinatal lethal with reduced brain levels of prostaglandin E2 The non-functional phospholipase A2-activating protein and the associated neurological phenotype, reported herein for the first time, join other complex phospholipid defects that cause leukoencephalopathies in humans, emphasizing the importance of this axis in white matter development and maintenance.


Subject(s)
Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Leukoencephalopathies/physiopathology , Proteins/genetics , Proteins/metabolism , Adolescent , Animals , Brain/embryology , Brain/growth & development , Brain/metabolism , Brain/pathology , Child , Consanguinity , Dinoprostone/metabolism , Embryo, Mammalian , Family Health , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Gene Expression Regulation/genetics , Humans , Leukoencephalopathies/diagnostic imaging , Lung/pathology , Male , Mice , Mice, Transgenic , Models, Molecular , NF-kappa B/metabolism , Phospholipases A2/metabolism , Skin/pathology
11.
J Cutan Pathol ; 45(2): 180-183, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29210482

ABSTRACT

Nevus spilus is a melanocytic neoplasm characterized by a tan macular background punctuated by multiple hyperpigmented macules or papules that represent various types of nevi. These include junctional and compound nevi, Spitz nevi, and rarely blue nevi. We report a unique case of widespread, multiple nevi spili giving rise to agminated Spitz nevi and congenital-pattern compound nevi. We performed genetic analysis to further characterize the mutational profile of this rare entity.


Subject(s)
Nevus, Epithelioid and Spindle Cell/genetics , Nevus, Pigmented/genetics , Skin Neoplasms/genetics , Adolescent , DNA Mutational Analysis , Female , Humans , Nevus, Epithelioid and Spindle Cell/congenital , Nevus, Epithelioid and Spindle Cell/pathology , Nevus, Pigmented/congenital , Nevus, Pigmented/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/congenital , Skin Neoplasms/pathology
12.
Proteomics ; 17(6)2017 03.
Article in English | MEDLINE | ID: mdl-28195392

ABSTRACT

Discovery proteomics experiments include many options for sample preparation and MS data acquisition, which are capable of creating datasets for quantifying thousands of proteins. To define a strategy that would produce a dataset with sufficient content while optimizing required resources, we compared (1) single-sample LC-MS/MS with data-dependent acquisition to single-sample LC-MS/MS with data-independent acquisition and (2) peptide fractionation with label-free (LF) quantification to peptide fractionation with relative quantification of chemically labeled peptides (sixplex tandem mass tags (TMT)). These strategies were applied to the same set of four frozen lung squamous cell carcinomas and four adjacent tissues, and the overall outcomes of each experiment were assessed. We identified 6656 unique protein groups with LF, 5535 using TMT, 3409 proteins from single-sample analysis with data-independent acquisition, and 2219 proteins from single-sample analysis with data-dependent acquisition. Pathway analysis indicated the number of proteins per pathway was proportional to the total protein identifications from each method, suggesting limited biological bias between experiments. The results suggest the use of single-sample experiments as a rapid tissue assessment tool and digestion quality control or as a technique to maximize output from limited samples and use of TMT or LF quantification as methods for larger amounts of tumor tissue with the selection being driven mainly by instrument time limitations. Data are available via ProteomeXchange with identifiers PXD004682, PXD004683, PXD004684, and PXD005733.


Subject(s)
Chromatography, Liquid/methods , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Proteome/metabolism , Proteomics/methods , Tandem Mass Spectrometry/methods , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Humans , Peptides/metabolism , Staining and Labeling
13.
Am J Hum Genet ; 95(1): 66-76, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24975946

ABSTRACT

Coronary artery calcification (CAC) is a heritable and definitive morphologic marker of atherosclerosis that strongly predicts risk for future cardiovascular events. To search for genes involved in CAC, we used an integrative transcriptomic, genomic, and protein expression strategy by using next-generation DNA sequencing in the discovery phase with follow-up studies using traditional molecular biology and histopathology techniques. RNA sequencing of peripheral blood from a discovery set of CAC cases and controls was used to identify dysregulated genes, which were validated by ClinSeq and Framingham Heart Study data. Only a single gene, TREML4, was upregulated in CAC cases in both studies. Further examination showed that rs2803496 was a TREML4 cis-eQTL and that the minor allele at this locus conferred up to a 6.5-fold increased relative risk of CAC. We characterized human TREML4 and demonstrated by immunohistochemical techniques that it is localized in macrophages surrounding the necrotic core of coronary plaques complicated by calcification (but not in arteries with less advanced disease). Finally, we determined by von Kossa staining that TREML4 colocalizes with areas of microcalcification within coronary plaques. Overall, we present integrative RNA, DNA, and protein evidence implicating TREML4 in coronary artery calcification. Our findings connect multimodal genomics data with a commonly used clinical marker of cardiovascular disease.


Subject(s)
Calcinosis , Coronary Vessels/pathology , DNA/metabolism , Proteins/metabolism , RNA/metabolism , Receptors, Immunologic/physiology , Base Sequence , DNA Primers , HEK293 Cells , Humans , Quantitative Trait Loci , Receptors, Immunologic/genetics
14.
Oncologist ; 22(2): 144-151, 2017 02.
Article in English | MEDLINE | ID: mdl-28179575

ABSTRACT

BACKGROUND: The increasing practicality of genomic sequencing technology has led to its incorporation into routine clinical practice. Successful identification and targeting of driver genomic alterations that provide proliferative and survival advantages to tumor cells have led to approval and ongoing development of several targeted cancer therapies. Within many major cancer centers, molecular tumor boards are constituted to shepherd precision medicine into clinical practice. MATERIALS AND METHODS: In July 2014, the Clinical Genomics Action Committee (CGAC) was established as the molecular tumor board companion to the Personalized Medicine Clinical Service (PMCS) at Moffitt Cancer Center in Tampa, Florida. The processes and outcomes of the program were assessed in order to help others move into the practice of precision medicine. RESULTS: Through the establishment and initial 1,400 patients of the PMCS and its associated molecular tumor board at a major cancer center, five practical lessons of broad applicability have been learned: transdisciplinary engagement, the use of the molecular report as an aid to clinical management, clinical actionability, getting therapeutic options to patients, and financial considerations. Value to patients includes access to cutting-edge practice merged with individualized preferences in treatment and care. CONCLUSIONS: Genomic-driven cancer medicine is increasingly becoming a part of routine clinical practice. For successful implementation of precision cancer medicine, strategically organized molecular tumor boards are critical to provide objective evidence-based translation of observed molecular alterations into patient-centered clinical action. Molecular tumor board implementation models along with clinical and economic outcomes will define future treatment standards. The Oncologist 2017;22:144-151Implications for Practice: It is clear that the increasing practicality of genetic tumor sequencing technology has led to its incorporation as part of routine clinical practice. Subsequently, many cancer centers are seeking to develop a personalized medicine services and/or molecular tumor board to shepherd precision medicine into clinical practice. This article discusses the key lessons learned through the establishment and development of a molecular tumor board and personalized medicine clinical service. This article highlights practical issues and can serve as an important guide to other centers as they conceive and develop their own personalized medicine services and molecular tumor boards.


Subject(s)
Genomics , Molecular Targeted Therapy/methods , Neoplasms/therapy , Precision Medicine/methods , Female , Humans , Male
15.
BMC Cancer ; 17(1): 250, 2017 04 08.
Article in English | MEDLINE | ID: mdl-28390395

ABSTRACT

BACKGROUND: Myoepithelial carcinoma of soft tissue is a rare, malignant neoplasm that is morphologically and immunophenotypically similar to its counterpart in salivary gland. It demonstrates myoepithelial differentiation, possessing both epithelial and myogenic characteristics. Thought to be chemotherapy insensitive, the optimal treatment regimen of this tumor has yet to be established and only a select few cases in the literature discuss treatment efficacy in detail. CASE PRESENTATION: Here we present a case of a young adult with metastatic myoepithelial carcinoma with an initial excellent response to systemic therapy utilizing carboplatin and paclitaxel with continued complete response after 3Ā years. The patient also underwent complete surgical excision and received adjuvant radiation to the primary site of disease. Exome sequencing revealed an inactivating mutation in RB1 which we believe to be the first such mutation to be reported in this cancer type. CONCLUSIONS: Given increasing evidence suggesting RB1 loss is associated with responsiveness to conventional chemotherapies, particularly platinum-based regimens, we hypothesize that this genetic feature predisposed chemosensitivity in our patient's tumor.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mutation , Myoepithelioma/therapy , Retinoblastoma Binding Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Adult , Carboplatin/therapeutic use , Humans , Male , Myoepithelioma/genetics , Neoplasm Metastasis , Paclitaxel/therapeutic use , Radiotherapy, Adjuvant , Treatment Outcome
16.
Cancer ; 122(18): 2828-35, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27244218

ABSTRACT

BACKGROUND: The incidence and outcomes of patients with colorectal cancer (CRC) varies by age. Younger patients tend to have sporadic cancers that are not detected by screening and worse survival. To understand whether genetic differences exist between age cohorts, the authors sought to characterize unique genetic alterations in patients with CRC. METHODS: In total, 283 patients who were diagnosed with sporadic CRC between 1998 and 2010 were identified and divided by age into 2 cohorts-ages ≤45 years (the younger cohort) and ≥65 years (the older cohort)-and targeted exome sequencing was performed. The Fisher exact test was used to detect differences in mutation frequencies between the 2 groups. Whole exome sequencing was performed on 21 additional younger patient samples for validation. Findings were confirmed in The Cancer Genome Atlas CRC data set. RESULTS: In total, 246 samples were included for final analysis (195 from the older cohort and 51 from the younger cohort). Mutations in the FBXW7 gene were more common in the younger cohort (27.5% vs 9.7%; P = .0022) as were mutations in the proofreading domain of polymerase ƎĀµ catalytic subunit (POLE) (9.8% vs 1%; P = .0048). There were similar mutation rates between cohorts with regard to TP53 (64.7% vs 61.5%), KRAS (43.1% vs 46.2%), and APC (60.8% vs 73.8%). BRAF mutations were numerically more common in the older cohort, although the difference did not reach statistical significance (2% vs 9.7%; P = .082). CONCLUSIONS: In this retrospective study, a unique genetic profile was identified for younger patients who have CRC compared with patients who are diagnosed at an older age. These findings should be validated in a larger study and could have an impact on future screening and treatment modalities for younger patients with CRC. Cancer 2016. Ā© 2016 American Cancer Society. Cancer 2016;122:2828-2835. Ā© 2016 American Cancer Society.


Subject(s)
Cell Cycle Proteins/genetics , Colorectal Neoplasms/genetics , DNA Polymerase II/genetics , F-Box Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Age Factors , Aged , Aged, 80 and over , Cell Cycle Proteins/metabolism , Cohort Studies , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , DNA Polymerase II/metabolism , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Female , Humans , Incidence , Male , Middle Aged , Mutation , Poly-ADP-Ribose Binding Proteins , Retrospective Studies , Ubiquitin-Protein Ligases/metabolism
17.
Cancer Control ; 23(1): 61-6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27009459

ABSTRACT

BACKGROUND: Uterine carcinosarcoma, a rare gynecological malignancy, often presents at the advanced stage with a poor prognosis because current therapies have not improved rates of survival. Genetic characterization of this tumor may lead to novel, specifically targeted drug targets to provide better treatment options for patients with this malignancy. METHODS: We present a case of a woman aged 61 years with uterine carcinosarcoma and retrospectively analyzed 100 study patients with uterine carcinosarcoma. From this group, 9 study patients underwent targeted sequencing of 1,321 genes. RESULTS: All 9 study patients had at least 1 mutation in JAK2, KRAS, PIK3CA, CTNNB1, PTEN, FBXW7, TP53, and ERBB2; of these, TP53 was the most frequently mutated gene (6/9). In addition, ARID1A and KMT2C, which have been described and identified as part of a set of chromatin-remodeling genes, were also found in our analyses. From our 100-person cohort clinical analyses, study patients with stage 1 cancer had a median survival rate of 33 months (95% confidence interval, 19-109) compared with a median survival rate of 6 months (95% confidence interval, 3-12) in those with stage 4 disease. CONCLUSIONS: Disease stage alone predicted the rate of clinical survival. Up to 50% in the study group were identified at having early stage disease (stage 1/2), indicating improved rates of overall detection compared with previously reported data. Our mutational analysis findings add to the number of tumors in which these mutations have been found and suggest that chromatin-remodeling dysregulation may play a role in the tumorigenesis of carcinosarcoma.


Subject(s)
Carcinogenesis/genetics , Carcinosarcoma/genetics , Chromatin Assembly and Disassembly/genetics , Uterine Neoplasms/genetics , Aged , Carcinosarcoma/diagnostic imaging , Carcinosarcoma/pathology , Cohort Studies , DNA Mutational Analysis , Female , Genetic Testing , Humans , Middle Aged , Mutation , Neoplasm Staging , Positron-Emission Tomography , Retrospective Studies , Survival Analysis , Uterine Neoplasms/diagnostic imaging , Uterine Neoplasms/pathology
18.
Proc Natl Acad Sci U S A ; 110(33): 13481-6, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23901115

ABSTRACT

Synonymous mutations, which do not alter the protein sequence, have been shown to affect protein function [Sauna ZE, Kimchi-Sarfaty C (2011) Nat Rev Genet 12(10):683-691]. However, synonymous mutations are rarely investigated in the cancer genomics field. We used whole-genome and -exome sequencing to identify somatic mutations in 29 melanoma samples. Validation of one synonymous somatic mutation in BCL2L12 in 285 samples identified 12 cases that harbored the recurrent F17F mutation. This mutation led to increased BCL2L12 mRNA and protein levels because of differential targeting of WT and mutant BCL2L12 by hsa-miR-671-5p. Protein made from mutant BCL2L12 transcript bound p53, inhibited UV-induced apoptosis more efficiently than WT BCL2L12, and reduced endogenous p53 target gene transcription. This report shows selection of a recurrent somatic synonymous mutation in cancer. Our data indicate that silent alterations have a role to play in human cancer, emphasizing the importance of their investigation in future cancer genome studies.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation/genetics , Genome, Human/genetics , Melanoma/genetics , Muscle Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Base Sequence , Blotting, Western , DNA Primers/genetics , Exome/genetics , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunoprecipitation , Lentivirus , MicroRNAs/genetics , Molecular Sequence Data , Muscle Proteins/metabolism , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Suppressor Protein p53/metabolism
19.
Br J Cancer ; 113(12): 1735-43, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26554648

ABSTRACT

BACKGROUND: The cyclin-dependent kinase inhibitor 3 (CDKN3) has been perceived as a tumour suppressor. Paradoxically, CDKN3 is often overexpressed in human cancer. It was unclear if CDKN3 overexpression is linked to alternative splicing variants or mutations that produce dominant-negative CDKN3. METHODS: We analysed CDKN3 expression and its association with patient survival in three cohorts of lung adenocarcinoma. We also examined CDKN3 mutations in the Cancer Genome Atlas (TCGA) and the Moffitt Cancer Center's Total Cancer Care (TCC) projects. CDKN3 transcripts were further analysed in a panel of cell lines and lung adenocarcinoma tissues. CDKN3 mRNA and protein levels in different cell cycle phases were examined. RESULTS: CDKN3 is overexpressed in non small cell lung cancer. High CDKN3 expression is associated with poor overall survival in lung adenocarcinoma. Two CDKN3 transcripts were detected in all samples. These CDKN3 transcripts represent the full length CDKN3 mRNA and a normal transcript lacking exon 2, which encodes an out of frame 23-amino acid peptide with little homology to CDKN3. CDKN3 mutations were found to be very rare. CDKN3 mRNA and protein were elevated during the mitosis phase of cell cycle. CONCLUSIONS: CDKN3 overexpression is prognostic of poor overall survival in lung adenocarcinoma. CDKN3 overexpression in lung adenocarcinoma is not attributed to alternative splicing or mutation but is likely due to increased mitotic activity, arguing against CDKN3 as a tumour suppressor.


Subject(s)
Adenocarcinoma/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dual-Specificity Phosphatases/genetics , Lung Neoplasms/genetics , RNA, Messenger/genetics , Survival Analysis , Amino Acid Sequence , Cohort Studies , Cyclin-Dependent Kinase Inhibitor Proteins/chemistry , Dual-Specificity Phosphatases/chemistry , Humans , Molecular Sequence Data
20.
Am J Hum Genet ; 91(1): 97-108, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22703879

ABSTRACT

Genome- and exome-sequencing costs are continuing to fall, and many individuals are undergoing these assessments as research participants and patients. The issue of secondary (so-called incidental) findings in exome analysis is controversial, and data are needed on methods of detection and their frequency. We piloted secondary variant detection by analyzing exomes for mutations in cancer-susceptibility syndromes in subjects ascertained for atherosclerosis phenotypes. We performed exome sequencing on 572 ClinSeq participants, and in 37 genes, we interpreted variants that cause high-penetrance cancer syndromes by using an algorithm that filtered results on the basis of mutation type, quality, and frequency and that filtered mutation-database entries on the basis of defined categories of causation. We identified 454 sequence variants that differed from the human reference. Exclusions were made on the basis of sequence quality (26 variants) and high frequency in the cohort (77 variants) or dbSNP (17 variants), leaving 334 variants of potential clinical importance. These were further filtered on the basis of curation of literature reports. Seven participants, four of whom were of Ashkenazi Jewish descent and three of whom did not meet family-history-based referral criteria, had deleterious BRCA1 or BRCA2 mutations. One participant had a deleterious SDHC mutation, which causes paragangliomas. Exome sequencing, coupled with multidisciplinary interpretation, detected clinically important mutations in cancer-susceptibility genes; four of such mutations were in individuals without a significant family history of disease. We conclude that secondary variants of high clinical importance will be detected at an appreciable frequency in exomes, and we suggest that priority be given to the development of more efficient modes of interpretation with trials in larger patient groups.


Subject(s)
Exome , Genetic Predisposition to Disease , Genetic Variation , Neoplasms/genetics , Penetrance , Aged , Algorithms , Atherosclerosis/genetics , Female , Humans , Incidental Findings , Male , Middle Aged , Mutation , Pedigree , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL