Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mod Pathol ; 36(11): 100294, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37532182

ABSTRACT

Gliomas harboring oncogenic ROS1 alterations are uncommon and primarily described in infants. Our goal was to characterize the clinicopathological features and molecular signatures of the full spectrum of ROS1 fusion-positive gliomas across all age groups. Through a retrospective multi-institutional collaboration, we report a collection of unpublished ROS1 fusion gliomas along with the characterization and meta-analysis of new and published cases. A cohort of 32 new and 58 published cases was divided into the following 3 age groups: 19 infants, 40 pediatric patients, and 31 adults with gliomas. Tumors in infants and adults showed uniformly high-grade morphology; however, tumors in pediatric patients exhibited diverse histologic features. The GOPC::ROS1 fusion was prevalent (61/79, 77%) across all age groups, and 10 other partner genes were identified. Adult tumors showed recurrent genomic alterations characteristic of IDH wild-type glioblastoma, including the +7/-10/CDKN2A deletion; amplification of CDK4, MDM2, and PDGFRA genes; and mutations involving TERTp, TP53, PIK3R1, PIK3CA, PTEN, and NF1 genes. Infant tumors showed few genomic alterations, whereas pediatric tumors showed moderate genomic complexity. The outcomes were significantly poorer in adult patients. Although not statistically significant, tumors in infant and pediatric patients with high-grade histology and in hemispheric locations appeared more aggressive than tumors with lower grade histology or those in nonhemispheric locations. In conclusion, this study is the largest to date to characterize the clinicopathological and molecular signatures of ROS1 fusion-positive gliomas from infant, pediatric, and adult patients. We conclude that ROS1 likely acts as a driver in infant and pediatric gliomas and as a driver or codriver in adult gliomas. Integrated comprehensive clinical testing might be helpful in identifying such patients for possible targeted therapy.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Child , Adult , Infant , Young Adult , Protein-Tyrosine Kinases/genetics , Retrospective Studies , Proto-Oncogene Proteins/genetics , Glioma/genetics , Glioma/pathology , Glioblastoma/genetics , Mutation , Brain Neoplasms/genetics , Brain Neoplasms/pathology
2.
J Mol Diagn ; 25(2): 87-93, 2023 02.
Article in English | MEDLINE | ID: mdl-36503148

ABSTRACT

Evaluation of suspected myeloid neoplasms involves testing for recurrent, diagnostically and therapeutically relevant genetic alterations. Current molecular testing requires multiple technologies, different domains of expertise, and unconnected workflows, resulting in variable, lengthy turnaround times that can delay treatment. To address this unmet clinical need, we evaluated the Oncomine Myeloid Assay GX panel on the Ion Torrent Genexus platform, a rapid, integrated nucleic acid to report next-generation sequencing platform for detecting clinically relevant genetic aberrations in myeloid disorders. Specimens included synthetic DNA (101 targets) and RNA (9 targets) controls and real-world nucleic acid material derived from bone marrow or peripheral blood samples (40 patients). Ion Torrent Genexus results and performance indices were compared with those obtained from clinically validated genomic testing workflows in 2 separate clinical laboratories. The Ion Torrent Genexus identified 100% of DNA and RNA control variants. For primary patient specimens, the Ion Torrent Genexus reported 82 of 107 DNA variants and 19 of 19 RNA gene fusions identified on clinically validated assays, yielding an 80% overall detection rate. Reanalysis of exported, unfiltered Ion Torrent Genexus data revealed 15 DNA variants not called by the filtered on-board bioinformatics pipeline, yielding a 92% potential detection rate. These results hold promise for the implementation of an integrated next-generation sequencing system to rapidly detect genetic aberrations, facilitating accurate, genomics-based diagnoses and accelerated time to precision therapies in myeloid neoplasms.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Humans , RNA/genetics , Mutation , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Neoplasms/genetics , High-Throughput Nucleotide Sequencing/methods , DNA/genetics , Semiconductors
3.
Cancer Genet ; 262-263: 5-15, 2022 04.
Article in English | MEDLINE | ID: mdl-34972036

ABSTRACT

The neurotrophic receptor tyrosine kinase (NTRK) family of genes, including NTRK1, NTRK2, and NTRK3, encodes membrane-bound receptors that normally regulate cell survival and differentiation upon binding of growth factors. Not surprisingly, mutations in these genes are known to contribute to the growth of a diverse number of cancers. With the recent FDA approval of two first-generation tyrosine-kinase inhibitors (TKIs) for adult and pediatric patients with solid tumors harboring NTRK gene fusions, much of the literature has focused on the biology behind these types of NTRK abnormalities; however, point mutations can also contribute to oncogenesis or resistance to TKI therapy, albeit at a lower frequency than fusions. This review focuses on NTRK gene mutations that are associated with resistance to directed therapies, mutations detected in the primary setting that confer increased oncogenic activity, and evidence that suggests that some of these variants may be treated using specific targeted therapies. Finally, this review focuses on the detection of point mutations, including the utility of cell-free DNA (cfDNA) for monitoring the acquisition of resistance mutations.


Subject(s)
Neoplasms , Point Mutation , Adult , Child , Humans , Membrane Glycoproteins/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Oncogenes , Receptor, trkA/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics
5.
J Mol Diagn ; 23(11): 1545-1552, 2021 11.
Article in English | MEDLINE | ID: mdl-34454115

ABSTRACT

Circulating cell-free DNA (ccfDNA) is used increasingly as a cancer biomarker for prognostication, as a correlate for tumor volume, or as input for downstream molecular analysis. Determining optimal blood processing and ccfDNA quantification are crucial for ccfDNA to serve as an accurate biomarker as it moves into the clinical realm. Whole blood was collected from 50 subjects, processed to plasma, and used immediately or frozen at -80°C. Plasma ccfDNA was extracted and concentration was assessed by real-time quantitative PCR (qPCR), fluorimetry, and droplet digital PCR (ddPCR). For the 24 plasma samples from metastatic pancreatic cancer patients, the variant allele fractions (VAF) of KRAS G12/13 pathogenic variants in circulating tumor DNA (ctDNA) were measured by ddPCR. Using a high-speed (16,000 × g) or slower-speed (4100 × g) second centrifugation step showed no difference in ccfDNA yield or ctDNA VAF. A two- versus three-spin centrifugation protocol also showed no difference in ccfDNA yield or ctDNA VAF. A higher yield was observed from fresh versus frozen plasma by qPCR and fluorimetry, whereas a higher yield was observed for frozen versus fresh plasma by ddPCR, however, no difference was observed in ctDNA VAF. Overall, our findings suggest factors to consider when implementing a ccfDNA extraction and quantification workflow in a research or clinical setting.


Subject(s)
Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/genetics , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Molecular Diagnostic Techniques/methods , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/genetics , Real-Time Polymerase Chain Reaction/methods , Alleles , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Blood Specimen Collection/methods , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Circulating Tumor DNA/isolation & purification , Cohort Studies , Humans , Mutation , Neoplasm Metastasis , Pancreatic Neoplasms/pathology
6.
Cancer Genet ; 256-257: 68-76, 2021 08.
Article in English | MEDLINE | ID: mdl-33915454

ABSTRACT

Acute myeloid leukemia (AML) is typically characterized clinically for prognostic purposes using both cytogenetic and molecular characteristics. However, both cytogenetic and molecular risk stratification schemas are varied and few reports have studied correlations between these schemas. We have performed a single institution retrospective review of cytogenetic and molecular classifications of AMLs seen at Penn Medicine between 2013 and 2018. One-hundred fourty-four cases were characterized according to European Leukemia Net (ELN) or Medical Research Council (MRC) criteria for cytogenetics and results compared to molecular profiling. When we analyzed the most common sequencing study results within the risk groupings, negative sequencing studies and FLT3 mutations were common in favorable AMLs, intermediate AMLs had mutations in FLT3, NPM1, DNMT3A and IDH2, while adverse AMLs had a high prevalence of TP53 mutations. We next grouped the genes on the panel by their proteins' functions and found mutations in signaling pathway genes to be common in favorable AMLs while tumor suppressors were commonly mutated in adverse AMLs. AMLs grouped by the type of chromosomal abnormality present showed that FLT3 mutations were common in AMLs with a trisomy while TP53 mutations were common in AMLs with a monosomy or a deletion. TP53 mutations are especially common in AMLs with a monosomal karyotype and often overlap with 17p loss. Interestingly, although all AMLs with TP53 mutations have a defect in the response to DNA damage, expression of P53 protein before and after irradiation is not consistently predicted by phenotype. Overall, these studies confirm the genetic complexity of AML which does not fall into simple patterns of cooperating mutations.


Subject(s)
Cytogenetic Analysis , Gene Expression Profiling , Leukemia, Myeloid, Acute/genetics , Cell Line, Tumor , Chromosome Aberrations , Cohort Studies , Gamma Rays , Humans , Mutation/genetics , Nucleophosmin , Risk Assessment , Risk Factors , Tumor Suppressor Protein p53/genetics
7.
Mod Pathol ; 34(5): 904-921, 2021 05.
Article in English | MEDLINE | ID: mdl-33311649

ABSTRACT

Massively parallel sequencing (MPS) has become a viable diagnostic tool to interrogate genetic profiles of numerous tumors but has yet to be routinely adopted in the setting of lymphoma. Here, we report the empirical application of a targeted 40-gene panel developed for use in mature lymphoid neoplasms (MLNs) and report our experience on over 500 cases submitted for MPS during the first year of its clinical use. MPS was applied to both fresh and fixed specimens. The most frequent diagnoses were diffuse large B-cell lymphoma (116), chronic lymphocytic leukemia/small lymphocytic lymphoma (60), marginal zone lymphoma (52), and follicular lymphoma (43), followed by a spectrum of mature T-cell neoplasms (40). Of 534 cases submitted, 471 generated reportable results in MLNs, with disease-associated variants (DAVs) detected in 241 cases (51.2%). The most frequent DAVs affected TP53 (30%), CREBBP (14%), MYD88 (14%), TNFRSF14 (10%), TNFAIP3 (10%), B2M (7%), and NOTCH2 (7%). The bulk of our findings confirm what is reported in the scientific literature. While a substantial majority of mutations did not directly impact diagnosis, MPS results were utilized to either change, refine, or facilitate the final diagnosis in ~10.8% of cases with DAVs and 5.5% of cases overall. In addition, we identified preanalytic variables that significantly affect assay performance highlighting items for specimen triage. We demonstrate the technical viability and utility of the judicious use of a targeted MPS panel that may help to establish general guidelines for specimen selection and diagnostic application in MLNs in routine clinical practice.


Subject(s)
Biomarkers, Tumor/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Lymphoma, B-Cell, Marginal Zone/diagnosis , Lymphoma, Follicular/diagnosis , Lymphoma, Large B-Cell, Diffuse/diagnosis , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, Follicular/genetics , Lymphoma, Large B-Cell, Diffuse/genetics
8.
Am J Surg Pathol ; 44(11): 1535-1540, 2020 11.
Article in English | MEDLINE | ID: mdl-32773530

ABSTRACT

Primary pulmonary myxoid sarcoma (PPMS) is a recently reported, exceedingly rare low-grade lung neoplasm characterized by reticular/lace-like growth of spindle to epithelioid cells embedded in an abundant myxoid matrix. Morphologically, it overlaps with a myxoid variant of angiomatoid fibrous histiocytoma (AFH) of the soft tissue. Genetically, they were both reported to harbor EWSR1-CREB1 fusion, while EWSR1-ATF1 has only been reported in AFH thus far. We report a case of primary pulmonary low-grade myxoid spindle cell tumor with morphologic and immunohistochemical features of PPMS but with an EWSR1-ATF1 fusion gene. In addition, we also encountered a case of endobronchial AFH with EWSR1-CREB1 translocation but also focal morphologic features of PPMS. These findings provide new evidence supporting the concept that PPMS and a myxoid variant of AFH represent a continuum with overlapping histologic, immunohistochemical, and genetic features.


Subject(s)
Histiocytoma, Malignant Fibrous/genetics , Histiocytoma, Malignant Fibrous/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Myxosarcoma/genetics , Myxosarcoma/pathology , Oncogene Proteins, Fusion/genetics , Adult , Humans , Male , Middle Aged
9.
Sci Data ; 7(1): 116, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286315

ABSTRACT

Understanding the aberrant transcriptional landscape of neuroblastoma is necessary to provide insight to the underlying influences of the initiation, progression and persistence of this developmental cancer. Here, we present chromatin immunoprecipitation sequencing (ChIP-Seq) data for the oncogenic transcription factors, MYCN and MYC, as well as regulatory histone marks H3K4me1, H3K4me3, H3K27Ac, and H3K27me3 in ten commonly used human neuroblastoma-derived cell line models. In addition, for all of the profiled cell lines we provide ATAC-Seq as a measure of open chromatin. We validate specificity of global MYCN occupancy in MYCN amplified cell lines and functional redundancy of MYC occupancy in MYCN non-amplified cell lines. Finally, we show with H3K27Ac ChIP-Seq that these cell lines retain expression of key neuroblastoma super-enhancers (SE). We anticipate this dataset, coupled with available transcriptomic profiling on the same cell lines, will enable the discovery of novel gene regulatory mechanisms in neuroblastoma.


Subject(s)
Epigenomics , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Cell Line, Tumor , Chromatin/genetics , Chromatin Immunoprecipitation , Gene Expression Profiling , Histones/genetics , Humans
10.
Front Oncol ; 10: 302, 2020.
Article in English | MEDLINE | ID: mdl-32211329

ABSTRACT

We developed a computational pipeline designed to use RNA sequencing (n = 136) and gene expression profiling (n = 250) data from neuroblastoma tumors to identify cell surface proteins predicted to be highly expressed in MYCN amplified neuroblastomas and with little or no expression in normal human tissues. We then performed ChIP-seq in the MYCN amplified cell lines KELLY, NB-1643, and NGP to identify gene promoters that are occupied by MYCN protein to define the intersection with the differentially-expressed gene list. We initially identified 116 putative immunotherapy targets with predicted transmembrane domains, with the most significant differentially-expressed of these being the calmodulin kinase-like vesicle-associated gene (CAMKV, p = 2 × 10-6). CAMKV encodes a protein that binds calmodulin in the presence of calcium, but lacks the kinase activity of other calmodulin kinase family members. We confirmed that CAMKV is selectively expressed in 7/7 MYCN amplified neuroblastoma cell lines and showed that the transcription of CAMKV is directly controlled by MYCN. From membrane fractionation and immunohistochemistry, we verified that CAMKV is membranous in MYCN amplified neuroblastoma cell lines and patient-derived xenografts. Finally, immunohistochemistry showed that CAMKV is not expressed on normal tissues outside of the central nervous system. Together, these data demonstrate that CAMKV is a differentially-expressed cell surface protein that is transcriptionally regulated by MYCN, making it a candidate for targeting with antibodies or antibody-drug conjugates that do not cross the blood brain barrier.

11.
Acta Cytol ; 64(1-2): 147-154, 2020.
Article in English | MEDLINE | ID: mdl-30995656

ABSTRACT

As the value of molecular testing of cancer specimens increases, the number of tests imposed on tumor specimens also increases, often in tension with the amount of tumor material available. To develop and validate molecular assays for limited specimens, there are specific concerns that must be addressed, including DNA quality, quantity, and abundance; the number of targets/ability to multiplex; and the analytical sensitivity and specificity of the assay itself. Ultimately, weighing these considerations during assay validation in the overall context of clinical utility and laboratory workflow is critical for delivering the highest level of personalized care to patients.


Subject(s)
Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , High-Throughput Nucleotide Sequencing/methods , Mutation , Neoplasms/diagnosis , DNA, Neoplasm/genetics , Humans , Neoplasms/genetics , Reproducibility of Results , Sensitivity and Specificity
12.
Arch Pathol Lab Med ; 144(1): 90-98, 2020 01.
Article in English | MEDLINE | ID: mdl-31211614

ABSTRACT

CONTEXT.­: Next-generation sequencing is a high-throughput method for detecting genetic abnormalities and providing prognostic and therapeutic information for patients with cancer. Oncogenic fusion transcripts are among the various classifications of genetic abnormalities present in tumors and are typically detected clinically with fluorescence in situ hybridization (FISH). However, FISH probes only exist for a limited number of targets, do not provide any information about fusion partners, cannot be multiplex, and have been shown to be limited in specificity for common targets such as ALK. OBJECTIVE.­: To validate an anchored multiplex polymerase chain reaction-based panel for the detection of fusion transcripts in a university hospital-based clinical molecular diagnostics laboratory. DESIGN.­: We used 109 unique clinical specimens to validate a custom panel targeting 104 exon boundaries from 17 genes involved in fusions in solid tumors. The panel can accept as little as 100 ng of total nucleic acid from PreservCyt-fixed tissue, and formalin-fixed, paraffin-embedded specimens with as little as 10% tumor nuclei. RESULTS.­: Using FISH as the gold standard, this assay has a sensitivity of 88.46% and a specificity of 95.83% for the detection of fusion transcripts involving ALK, RET, and ROS1 in lung adenocarcinomas. Using a validated next-generation sequencing assay as the orthogonal gold standard for the detection of EGFR variant III (EGFRvIII) in glioblastomas, the assay is 92.31% sensitive and 100% specific. CONCLUSIONS.­: This multiplexed assay is tumor and fusion partner agnostic and will provide clinical utility in therapy selection for patients with solid tumors.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Oncogene Proteins, Fusion/analysis , Sequence Analysis, RNA/methods , Humans , Multiplex Polymerase Chain Reaction/methods , Protein Isoforms/analysis
13.
Cancer Genet ; 228-229: 55-63, 2018 12.
Article in English | MEDLINE | ID: mdl-30553474

ABSTRACT

One caveat of next-generation sequencing (NGS)-based clinical oncology testing is the high amount of input DNA required. We sought to develop a focused NGS panel that could capture hotspot regions in relevant genes requiring 0.5-10 ng input DNA. The resulting Penn Precision Panel (PPP) targeted 20 genes containing clinically significant variants relevant to many cancers. One hundred twenty-three samples were analyzed, including 83 solid tumor specimens derived from FFPE. Various input quantities of DNA (0.5-10 ng) were amplified with content-specific PCR primer pools, then sequenced on a MiSeq instrument (Illumina, Inc.) via paired-end, 2 × 186 base pair reads to an average read depth of greater than 6500x. Variants were detected using an in-house analysis pipeline. Clinical sensitivity and specificity were assessed using results from our previously validated solid tumor NGS panel; sensitivity of the PPP is 96.75% (387/400 variants) and specificity is 99.9% (8427/8428 base pairs). Variant allele frequencies (VAFs) are highly concordant across both assays (r = 0.98 p < 0.0001). The PPP is a robust, clinically validated test optimized for low-yield solid tumor specimens, capturing a high percentage of clinically relevant variants found by larger commercially available NGS panels while using only 0.5-10 ng of input DNA.


Subject(s)
DNA, Neoplasm/genetics , High-Throughput Nucleotide Sequencing/methods , DNA, Neoplasm/analysis , Humans , Limit of Detection
14.
Cancer Cell ; 32(3): 295-309.e12, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28898695

ABSTRACT

We developed an RNA-sequencing-based pipeline to discover differentially expressed cell-surface molecules in neuroblastoma that meet criteria for optimal immunotherapeutic target safety and efficacy. Here, we show that GPC2 is a strong candidate immunotherapeutic target in this childhood cancer. We demonstrate high GPC2 expression in neuroblastoma due to MYCN transcriptional activation and/or somatic gain of the GPC2 locus. We confirm GPC2 to be highly expressed on most neuroblastomas, but not detectable at appreciable levels in normal childhood tissues. In addition, we demonstrate that GPC2 is required for neuroblastoma proliferation. Finally, we develop a GPC2-directed antibody-drug conjugate that is potently cytotoxic to GPC2-expressing neuroblastoma cells. Collectively, these findings validate GPC2 as a non-mutated neuroblastoma oncoprotein and candidate immunotherapeutic target.


Subject(s)
Glypicans/metabolism , Immunotherapy , Molecular Targeted Therapy , Neuroblastoma/immunology , Neuroblastoma/therapy , Oncogene Proteins/metabolism , Animals , Antibodies, Neoplasm/metabolism , Cell Death , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Child , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Mice, Inbred NOD , Mice, SCID , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors
15.
Mol Cell ; 66(2): 247-257.e5, 2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28410996

ABSTRACT

Recruitment of transcription factors (TFs) to repressed genes in euchromatin is essential to activate new transcriptional programs during cell differentiation. However, recruitment of all TFs, including pioneer factors, is impeded by condensed H3K27me3-containing chromatin. Single-cell and gene-specific analyses revealed that, during the first hours of induction of differentiation of mammalian embryonic stem cells (ESCs), accumulation of the repressive histone mark H3K27me3 is delayed after DNA replication, indicative of a decondensed chromatin structure in all regions of the replicating genome. This delay provides a critical "window of opportunity" for recruitment of lineage-specific TFs to DNA. Increasing the levels of post-replicative H3K27me3 or preventing S phase entry inhibited recruitment of new TFs to DNA and significantly blocked cell differentiation. These findings suggest that recruitment of lineage-specifying TFs occurs soon after replication and is facilitated by a decondensed chromatin structure. This insight may explain the developmental plasticity of stem cells and facilitate their exploitation for therapeutic purposes.


Subject(s)
Cell Differentiation , Cell Lineage , Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA Replication , DNA/biosynthesis , Embryonic Stem Cells/metabolism , Histones/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , Cell Plasticity , Chromatin/chemistry , DNA/chemistry , DNA/genetics , DNA Methylation , Gene Expression Regulation, Developmental , Histone Demethylases/metabolism , Histones/chemistry , Humans , Methylation , Mice , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Binding , Structure-Activity Relationship , Time Factors , Transcription Factors/genetics
16.
MAbs ; 8(4): 799-810, 2016.
Article in English | MEDLINE | ID: mdl-26910291

ABSTRACT

CD56 (NCAM, neural cell adhesion molecule) is over-expressed in many tumor types, including neuroblastoma, multiple myeloma, small cell lung cancer, ovarian cancer, acute myeloid leukemia, NK-T lymphoma, neuroendocrine cancer and pancreatic cancer. Using phage display, we identified 2 high-affinity anti-CD56 human monoclonal antibodies (mAbs), m900 and m906, which bound to spatially separated non-overlapping epitopes with similar affinity (equilibrium dissociation constant 2.9 and 4.5 nM, respectively). m900 bound to the membrane proximal fibronectin type III-like domains, whereas m906 bound to the N-terminal IgG-like domains. m906 induced significant down-regulation of CD56 in 4 neuroblastoma cell lines tested, while m900-induced downregulation of CD56 was much lower. Antibody-drug conjugates (ADCs) made by conjugation with a highly potent pyrrolobenzodiazepine dimer (PBD) exhibited killing activity that correlated with CD56 down-regulation, and to some extent with in vivo binding ability of the antibodies. The m906PBD ADC was much more potent than m900PBD, likely due to higher CD56-mediated downregulation and stronger binding to cells. Treatment with m906PBD ADC resulted in very potent cytotoxicity (IC50: 0.05-1.7 pM). These results suggest a novel approach for targeting CD56-expressing neuroblastoma cells. Further studies in animal models and in humans are needed to find whether these antibodies and their drug conjugates are promising candidate therapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , CD56 Antigen/immunology , Immunoconjugates/pharmacology , Neuroblastoma/immunology , Animals , Antibody Affinity , Cell Line, Tumor , Cell Survival/drug effects , Epitopes , Humans , Xenograft Model Antitumor Assays
17.
Nature ; 528(7582): 418-21, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26560027

ABSTRACT

Neuroblastoma is a paediatric malignancy that typically arises in early childhood, and is derived from the developing sympathetic nervous system. Clinical phenotypes range from localized tumours with excellent outcomes to widely metastatic disease in which long-term survival is approximately 40% despite intensive therapy. A previous genome-wide association study identified common polymorphisms at the LMO1 gene locus that are highly associated with neuroblastoma susceptibility and oncogenic addiction to LMO1 in the tumour cells. Here we investigate the causal DNA variant at this locus and the mechanism by which it leads to neuroblastoma tumorigenesis. We first imputed all possible genotypes across the LMO1 locus and then mapped highly associated single nucleotide polymorphism (SNPs) to areas of chromatin accessibility, evolutionary conservation and transcription factor binding sites. We show that SNP rs2168101 G>T is the most highly associated variant (combined P = 7.47 × 10(-29), odds ratio 0.65, 95% confidence interval 0.60-0.70), and resides in a super-enhancer defined by extensive acetylation of histone H3 lysine 27 within the first intron of LMO1. The ancestral G allele that is associated with tumour formation resides in a conserved GATA transcription factor binding motif. We show that the newly evolved protective TATA allele is associated with decreased total LMO1 expression (P = 0.028) in neuroblastoma primary tumours, and ablates GATA3 binding (P < 0.0001). We demonstrate allelic imbalance favouring the G-containing strand in tumours heterozygous for this SNP, as demonstrated both by RNA sequencing (P < 0.0001) and reporter assays (P = 0.002). These findings indicate that a recently evolved polymorphism within a super-enhancer element in the first intron of LMO1 influences neuroblastoma susceptibility through differential GATA transcription factor binding and direct modulation of LMO1 expression in cis, and this leads to an oncogenic dependency in tumour cells.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Genetic Predisposition to Disease/genetics , LIM Domain Proteins/genetics , Neuroblastoma/genetics , Polymorphism, Single Nucleotide/genetics , Transcription Factors/genetics , Acetylation , Alleles , Allelic Imbalance , Binding Sites , Epigenomics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genome-Wide Association Study , Genotype , Histones/chemistry , Histones/metabolism , Humans , Introns/genetics , Lysine/metabolism , Organ Specificity , Reproducibility of Results
18.
Cancer Discov ; 5(12): 1282-95, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26516065

ABSTRACT

UNLABELLED: The CD19 antigen, expressed on most B-cell acute lymphoblastic leukemias (B-ALL), can be targeted with chimeric antigen receptor-armed T cells (CART-19), but relapses with epitope loss occur in 10% to 20% of pediatric responders. We detected hemizygous deletions spanning the CD19 locus and de novo frameshift and missense mutations in exon 2 of CD19 in some relapse samples. However, we also discovered alternatively spliced CD19 mRNA species, including one lacking exon 2. Pull-down/siRNA experiments identified SRSF3 as a splicing factor involved in exon 2 retention, and its levels were lower in relapsed B-ALL. Using genome editing, we demonstrated that exon 2 skipping bypasses exon 2 mutations in B-ALL cells and allows expression of the N-terminally truncated CD19 variant, which fails to trigger killing by CART-19 but partly rescues defects associated with CD19 loss. Thus, this mechanism of resistance is based on a combination of deleterious mutations and ensuing selection for alternatively spliced RNA isoforms. SIGNIFICANCE: CART-19 yield 70% response rates in patients with B-ALL, but also produce escape variants. We discovered that the underlying mechanism is the selection for preexisting alternatively spliced CD19 isoforms with the compromised CART-19 epitope. This mechanism suggests a possibility of targeting alternative CD19 ectodomains, which could improve survival of patients with B-cell neoplasms.


Subject(s)
Alternative Splicing , Antigens, CD19/genetics , Immunotherapy , Mutation , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Antigens, CD19/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Membrane/metabolism , Epitopes/immunology , Exons , Humans , Immunotherapy/methods , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Protein Binding , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/metabolism , Recurrence , Sequence Analysis, DNA , Serine-Arginine Splicing Factors , Transcription, Genetic
19.
Cancer Res ; 74(1): 272-86, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24197134

ABSTRACT

Increasing evidence links deregulation of the ubiquitin-specific proteases 22 (USP22) deubitiquitylase to cancer development and progression in a select group of tumor types, but its specificity and underlying mechanisms of action are not well defined. Here we show that USP22 is a critical promoter of lethal tumor phenotypes that acts by modulating nuclear receptor and oncogenic signaling. In multiple xenograft models of human cancer, modeling of tumor-associated USP22 deregulation demonstrated that USP22 controls androgen receptor accumulation and signaling, and that it enhances expression of critical target genes coregulated by androgen receptor and MYC. USP22 not only reprogrammed androgen receptor function, but was sufficient to induce the transition to therapeutic resistance. Notably, in vivo depletion experiments revealed that USP22 is critical to maintain phenotypes associated with end-stage disease. This was a significant finding given clinical evidence that USP22 is highly deregulated in tumors, which have achieved therapeutic resistance. Taken together, our findings define USP22 as a critical effector of tumor progression, which drives lethal phenotypes, rationalizing this enzyme as an appealing therapeutic target to treat advanced disease.


Subject(s)
Biomarkers, Tumor/metabolism , Prostatic Neoplasms, Castration-Resistant/enzymology , Thiolester Hydrolases/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Androgen Receptor Antagonists/pharmacology , Animals , Cell Culture Techniques , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Gene Expression , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Receptors, Androgen/metabolism , Thiolester Hydrolases/deficiency , Thiolester Hydrolases/genetics , Ubiquitin Thiolesterase
20.
J Biol Chem ; 288(33): 24234-46, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23760504

ABSTRACT

Pluripotent embryonic stem cells (ESCs) undergo self-renewal until stimulated to differentiate along specific lineage pathways. Many of the transcriptional networks that drive reprogramming of a self-renewing ESC to a differentiating cell have been identified. However, fundamental questions remain unanswered about the epigenetic programs that control these changes in gene expression. Here we report that the histone ubiquitin hydrolase ubiquitin-specific protease 22 (USP22) is a critical epigenetic modifier that controls this transition from self-renewal to differentiation. USP22 is induced as ESCs differentiate and is necessary for differentiation into all three germ layers. We further report that USP22 is a transcriptional repressor of the locus encoding the core pluripotency factor sex-determining region Y-box 2 (SOX2) in ESCs, and this repression is required for efficient differentiation. USP22 occupies the Sox2 promoter and hydrolyzes monoubiquitin from ubiquitylated histone H2B and blocks transcription of the Sox2 locus. Our study reveals an epigenetic mechanism that represses the core pluripotency transcriptional network in ESCs, allowing ESCs to transition from a state of self-renewal into lineage-specific differentiation programs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endopeptidases/metabolism , Epigenesis, Genetic , SOXB1 Transcription Factors/genetics , Transcription, Genetic , Ubiquitin-Specific Proteases/metabolism , Animals , Cell Line , Cell Proliferation , Endopeptidases/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Loci/genetics , Histones/metabolism , Mice , Phenotype , Pluripotent Stem Cells/metabolism , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism , Sirtuin 1/metabolism , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases/genetics , Ubiquitination/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...