Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Nat Commun ; 15(1): 3732, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702309

ABSTRACT

Immunotherapy with chimeric antigen receptor T cells for pediatric solid and brain tumors is constrained by available targetable antigens. Cancer-specific exons present a promising reservoir of targets; however, these have not been explored and validated systematically in a pan-cancer fashion. To identify cancer specific exon targets, here we analyze 1532 RNA-seq datasets from 16 types of pediatric solid and brain tumors for comparison with normal tissues using a newly developed workflow. We find 2933 exons in 157 genes encoding proteins of the surfaceome or matrisome with high cancer specificity either at the gene (n = 148) or the alternatively spliced isoform (n = 9) level. Expression of selected alternatively spliced targets, including the EDB domain of fibronectin 1, and gene targets, such as COL11A1, are validated in pediatric patient derived xenograft tumors. We generate T cells expressing chimeric antigen receptors specific for the EDB domain or COL11A1 and demonstrate that these have antitumor activity. The full target list, explorable via an interactive web portal ( https://cseminer.stjude.org/ ), provides a rich resource for developing immunotherapy of pediatric solid and brain tumors using gene or AS targets with high expression specificity in cancer.


Subject(s)
Brain Neoplasms , Exons , Receptors, Chimeric Antigen , Humans , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Brain Neoplasms/genetics , Animals , Exons/genetics , Child , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Mice , Immunotherapy/methods , Alternative Splicing , Fibronectins/genetics , Fibronectins/metabolism , Fibronectins/immunology , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic , RNA-Seq , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor , Immunotherapy, Adoptive/methods
2.
Cell Rep Med ; 5(2): 101422, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38350450

ABSTRACT

The emergence of immune escape is a significant roadblock to developing effective chimeric antigen receptor (CAR) T cell therapies against hematological malignancies, including acute myeloid leukemia (AML). Here, we demonstrate feasibility of targeting two antigens simultaneously by combining a GRP78-specific peptide antigen recognition domain with a CD123-specific scFv to generate a peptide-scFv bispecific antigen recognition domain (78.123). To achieve this, we test linkers with varying length and flexibility and perform immunophenotypic and functional characterization. We demonstrate that bispecific CAR T cells successfully recognize and kill tumor cells that express GRP78, CD123, or both antigens and have improved antitumor activity compared to their monospecific counterparts when both antigens are expressed. Protein structure prediction suggests that linker length and compactness influence the functionality of the generated bispecific CARs. Thus, we present a bispecific CAR design strategy to prevent immune escape in AML that can be extended to other peptide-scFv combinations.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , Humans , T-Lymphocytes , Interleukin-3 Receptor alpha Subunit/metabolism , Endoplasmic Reticulum Chaperone BiP , Receptors, Chimeric Antigen/metabolism , Leukemia, Myeloid, Acute/pathology
3.
Res Sq ; 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38260279

ABSTRACT

Immunotherapy with CAR T cells for pediatric solid and brain tumors is constrained by available targetable antigens. Cancer-specific exons (CSE) present a promising reservoir of targets; however, these have not been explored and validated systematically in a pan-cancer fashion. To identify CSE targets, we analyzed 1,532 RNA-seq datasets from 16 types of pediatric solid and brain tumors for comparison with normal tissues using a newly developed workflow. We found 2,933 exons in 157 genes encoding proteins of the surfaceome or matrisome with high cancer specificity either at the gene (n=148) or the alternatively spliced (AS) isoform (n=9) level. Expression of selected AS targets, including the EDB domain of FN1 (EDB), and gene targets, such as COL11A1, were validated in pediatric PDX tumors. We generated CAR T cells specific to EDB or COL11A1 and demonstrated that COL11A1-CAR T-cells have potent antitumor activity. The full target list, explorable via an interactive web portal (https://cseminer.stjude.org/), provides a rich resource for developing immunotherapy of pediatric solid and brain tumors using gene or AS targets with high expression specificity in cancer.

4.
Cell Rep Med ; 4(11): 101297, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37992682

ABSTRACT

Lack of targetable antigens is a key limitation for developing successful T cell-based immunotherapies. Members of the unfolded protein response (UPR) represent ideal immunotherapy targets because the UPR regulates the ability of cancer cells to resist cell death, sustain proliferation, and metastasize. Glucose-regulated protein 78 (GRP78) is a key UPR regulator that is overexpressed and translocated to the cell surface of a wide variety of cancers in response to elevated endoplasmic reticulum (ER) stress. We show that GRP78 is highly expressed on the cell surface of multiple solid and brain tumors, making cell surface GRP78 a promising chimeric antigen receptor (CAR) T cell target. We demonstrate that GRP78-CAR T cells can recognize and kill GRP78+ brain and solid tumors in vitro and in vivo. Additionally, our findings demonstrate that GRP78 is upregulated on CAR T cells upon T cell activation; however, this expression is tumor-cell-line specific and results in heterogeneous GRP78-CAR T cell therapeutic response.


Subject(s)
Brain Neoplasms , Receptors, Chimeric Antigen , Humans , Endoplasmic Reticulum Chaperone BiP , Glucose , T-Lymphocytes , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Brain Neoplasms/therapy
5.
Nat Commun ; 14(1): 1739, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37019972

ABSTRACT

Oncogenic fusions formed through chromosomal rearrangements are hallmarks of childhood cancer that define cancer subtype, predict outcome, persist through treatment, and can be ideal therapeutic targets. However, mechanistic understanding of the etiology of oncogenic fusions remains elusive. Here we report a comprehensive detection of 272 oncogenic fusion gene pairs by using tumor transcriptome sequencing data from 5190 childhood cancer patients. We identify diverse factors, including translation frame, protein domain, splicing, and gene length, that shape the formation of oncogenic fusions. Our mathematical modeling reveals a strong link between differential selection pressure and clinical outcome in CBFB-MYH11. We discover 4 oncogenic fusions, including RUNX1-RUNX1T1, TCF3-PBX1, CBFA2T3-GLIS2, and KMT2A-AFDN, with promoter-hijacking-like features that may offer alternative strategies for therapeutic targeting. We uncover extensive alternative splicing in oncogenic fusions including KMT2A-MLLT3, KMT2A-MLLT10, C11orf95-RELA, NUP98-NSD1, KMT2A-AFDN and ETV6-RUNX1. We discover neo splice sites in 18 oncogenic fusion gene pairs and demonstrate that such splice sites confer therapeutic vulnerability for etiology-based genome editing. Our study reveals general principles on the etiology of oncogenic fusions in childhood cancer and suggests profound clinical implications including etiology-based risk stratification and genome-editing-based therapeutics.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Child , Core Binding Factor Alpha 2 Subunit/genetics , Oncogene Fusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcriptome , Causality , Oncogene Proteins, Fusion/genetics
6.
Cancer Cell ; 40(8): 809-811, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35944501

ABSTRACT

In this issue of Cancer Cell, Zaitsev et al. (2022) present a machine-learning-based approach, trained from millions of artificial transcriptomes with admixed cell populations, for reconstructing tumor microenvironments (TMEs). The high accuracy of this approach, demonstrated through extensive validation, enables systematic investigation of TMEs in both research and clinical settings.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Machine Learning , Neoplasms/genetics , Transcriptome , Tumor Microenvironment/genetics
7.
Int Urol Nephrol ; 54(11): 2901-2909, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35426589

ABSTRACT

BACKGROUND: Present investigation aims to elucidate safety and efficacy of hemodialysis as well as peritoneal dialysis in treating end-stage diabetic nephropathy. METHODS: We searched various databases for articles from the database starting date to October 2019. The analysis involved studies that contained outcomes of hemodialysis and peritoneal dialysis in the treatment of end-stage diabetic nephropathy. A total of 12 randomized controlled trials (RCTs) with 932 participants were collected. RESULTS: Meta-analysis results suggested that comparing with peritoneal dialysis group, hemodialysis group had a higher incidence of cardiovascular and cerebrovascular events and bleeding complications. There was no statistically significant difference regarding the infection (P = 0.71) or malnutrition (P = 0.53) incidence between the two forms of dialysis. Hemodialysis could better improve the levels of albumin [mean difference (MD) = 6.80, 95% CI = (4.17-9.44)] and hemoglobin [MD = 3.40, 95% CI = (0.94-5.86)] than peritoneal dialysis after 3 months or more. CONCLUSIONS: In treating end-stage diabetic nephropathy patients, peritoneal dialysis had a lower incidence of cardiovascular and cerebrovascular events, as well as bleeding complication than hemodialysis. However, hemodialysis could better improve albumin and hemoglobin levels than peritoneal dialysis after 3 months.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Kidney Failure, Chronic , Peritoneal Dialysis , Albumins , Diabetic Nephropathies/complications , Diabetic Nephropathies/therapy , Hemoglobins , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Peritoneal Dialysis/adverse effects , Peritoneal Dialysis/methods , Randomized Controlled Trials as Topic , Renal Dialysis/adverse effects , Renal Dialysis/methods
8.
Blood Cancer Discov ; 3(3): 194-207, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35176137

ABSTRACT

The genetics of relapsed pediatric acute myeloid leukemia (AML) has yet to be comprehensively defined. Here, we present the spectrum of genomic alterations in 136 relapsed pediatric AMLs. We identified recurrent exon 13 tandem duplications (TD) in upstream binding transcription factor (UBTF) in 9% of relapsed AML cases. UBTF-TD AMLs commonly have normal karyotype or trisomy 8 with cooccurring WT1 mutations or FLT3-ITD but not other known oncogenic fusions. These UBTF-TD events are stable during disease progression and are present in the founding clone. In addition, we observed that UBTF-TD AMLs account for approximately 4% of all de novo pediatric AMLs, are less common in adults, and are associated with poor outcomes and MRD positivity. Expression of UBTF-TD in primary hematopoietic cells is sufficient to enhance serial clonogenic activity and to drive a similar transcriptional program to UBTF-TD AMLs. Collectively, these clinical, genomic, and functional data establish UBTF-TD as a new recurrent mutation in AML. SIGNIFICANCE: We defined the spectrum of mutations in relapsed pediatric AML and identified UBTF-TDs as a new recurrent genetic alteration. These duplications are more common in children and define a group of AMLs with intermediate-risk cytogenetic abnormalities, FLT3-ITD and WT1 alterations, and are associated with poor outcomes. See related commentary by Hasserjian and Nardi, p. 173. This article is highlighted in the In This Issue feature, p. 171.


Subject(s)
Leukemia, Myeloid, Acute , Adult , Child , Chromosome Aberrations , Exons , Genomics , Humans , Leukemia, Myeloid, Acute/genetics , Mutation , Recurrence
9.
Biotechnol Appl Biochem ; 69(4): 1459-1473, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34159631

ABSTRACT

To investigate the properties of carotenoids from the extremophile Deinococcus xibeiensis R13, the factors affecting the stability of carotenoids extracted from D. xibeiensis R13, including temperature, illumination, pH, redox chemicals, metal ions, and food additives, were investigated. The results showed that low temperature, neutral pH, reducing agents, Mn2+ , and food additives (xylose and glucose) can effectively improve the stability of Deinococcus carotenoids. The carotenoids of D. xibeiensis R13 exhibited strong antioxidant activity, with the scavenging rate of hydroxyl radicals reaching 71.64%, which was higher than the scavenging efficiency for 1,1-diphenyl-2-picrylhydrazyl free radicals and 2,2'-azino-bis (3-ethyl-benzothiazoline-6-sulfonic acid) free radicals (44.55 and 27.65%, respectively). In addition, the total antioxidant capacity reached 0.60 U/ml, which was 2.61-fold that of carotenoids from the model strain Deinococcus radiodurans R1. Finally, we predicted the gene clusters encoding carotenoid biosynthesis pathways in the genome of R13 and identified putative homologous genes. The key enzyme genes (crtE, crtB, crtI, crtLm, cruF, crtD, and crtO) in carotenoid synthesis of D. xibeiensis R13 were cloned to construct the multigene coexpression plasmids pET-EBI and pRSF-LmFDO. The carotenoid biosynthesis pathway was heterologously introduced into engineered Escherichia coli EBILmFDO, which exhibited a higher yield (7.14 mg/L) than the original strain. These analysis results can help us to better understand the metabolic synthesis of carotenoids in extremophiles.


Subject(s)
Carotenoids , Deinococcus , Antioxidants/metabolism , Carotenoids/chemistry , Carotenoids/metabolism , Deinococcus/genetics , Deinococcus/metabolism , Food Additives , Free Radicals/metabolism
10.
BMC Cancer ; 21(1): 1233, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34789196

ABSTRACT

BACKGROUND: RNA editing leads to post-transcriptional variation in protein sequences and has important biological implications. We sought to elucidate the landscape of RNA editing events across pediatric cancers. METHODS: Using RNA-Seq data mapped by a pipeline designed to minimize mapping ambiguity, we investigated RNA editing in 711 pediatric cancers from the St. Jude/Washington University Pediatric Cancer Genome Project focusing on coding variants which can potentially increase protein sequence diversity. We combined de novo detection using paired tumor DNA-RNA data with analysis of known RNA editing sites. RESULTS: We identified 722 unique RNA editing sites in coding regions across pediatric cancers, 70% of which were nonsynonymous recoding variants. Nearly all editing sites represented the canonical A-to-I (n = 706) or C-to-U sites (n = 14). RNA editing was enriched in brain tumors compared to other cancers, including editing of glutamate receptors and ion channels involved in neurotransmitter signaling. RNA editing profiles of each pediatric cancer subtype resembled those of the corresponding normal tissue profiled by the Genotype-Tissue Expression (GTEx) project. CONCLUSIONS: In this first comprehensive analysis of RNA editing events in pediatric cancer, we found that the RNA editing profile of each cancer subtype is similar to its normal tissue of origin. Tumor-specific RNA editing events were not identified indicating that successful immunotherapeutic targeting of RNA-edited peptides in pediatric cancer should rely on increased antigen presentation on tumor cells compared to normal but not on tumor-specific RNA editing per se.


Subject(s)
Neoplasms/genetics , RNA Editing , Sequence Analysis, RNA/methods , Brain Neoplasms/genetics , Child , DNA, Neoplasm , Humans , Immunotherapy , Neoplasms/metabolism , Neoplasms/therapy , Open Reading Frames , Organ Specificity , RNA, Neoplasm , Whole Genome Sequencing
12.
Sci Rep ; 11(1): 5154, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664368

ABSTRACT

USP7, which encodes a deubiquitylating enzyme, is among the most frequently mutated genes in pediatric T-ALL, with somatic heterozygous loss-of-function mutations (haploinsufficiency) predominantly affecting the subgroup that has aberrant TAL1 oncogene activation. Network analysis of > 200 T-ALL transcriptomes linked USP7 haploinsufficiency with decreased activities of E-proteins. E-proteins are also negatively regulated by TAL1, leading to concerted down-regulation of E-protein target genes involved in T-cell development. In T-ALL cell lines, we showed the physical interaction of USP7 with E-proteins and TAL1 by mass spectrometry and ChIP-seq. Haploinsufficient but not complete CRISPR knock-out of USP7 showed accelerated cell growth and validated transcriptional down-regulation of E-protein targets. Our study unveiled the synergistic effect of USP7 haploinsufficiency with aberrant TAL1 activation on T-ALL, implicating USP7 as a haploinsufficient tumor suppressor in T-ALL. Our findings caution against a universal oncogene designation for USP7 while emphasizing the dosage-dependent consequences of USP7 inhibitors currently under development as potential cancer therapeutics.


Subject(s)
Oncogenes/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Ubiquitin-Specific Peptidase 7/genetics , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Lineage/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Leukemic/genetics , Haploinsufficiency/genetics , Humans , Pediatrics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcriptional Activation/genetics
13.
Bioprocess Biosyst Eng ; 44(6): 1033-1047, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33486569

ABSTRACT

To construct a Saccharomyces cerevisiae strain for efficient lycopene production, we used a pathway engineering strategy based on expression modules comprising fusion proteins and a strong constitutive promoter. The two recombinant plasmids pEBI encoding the fusion genes with an inducible promoter, as well as pIETB with a constitutive promoter and terminator were introduced into S. cerevisiae YPH499 and BY4741 to obtain the four recombinant strains ypEBI, ypIETB, byEBI and byIETB. The lycopene production and the transcription levels of key genes were higher in the BY4741 chassis than in YPH499. Accordingly, the content of total and unsaturated fatty acids was also higher in BY4741, which also exhibited a decrease of glucose, increase of trehalose, increase of metabolite in citrate cycle, and low levels of amino acids. These changes rerouted metabolic fluxes toward lycopene synthesis, indicating that the BY4741 chassis was more suitable for lycopene synthesis. The lycopene content of bpIETB in SG-Leu medium supplemented with 100 mg/L of linolenic acid reached 10.12 mg/g dry cell weight (DCW), which was 85.7% higher than without the addition of unsaturated fatty acids. The constitutive promoter expression strategy employed in this study achieved efficient lycopene synthesis in S. cerevisiae, and the strain bpIETB was obtained a suitable chassis host for lycopene production, which provides a basis for further optimization of lycopene production in artificial synthetic cells and a reference for the multi-enzyme synthesis of other similar complex terpenoids.


Subject(s)
Lycopene/metabolism , Metabolic Engineering , Metabolic Networks and Pathways , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
14.
Cancer Discov ; 11(5): 1082-1099, 2021 05.
Article in English | MEDLINE | ID: mdl-33408242

ABSTRACT

Effective data sharing is key to accelerating research to improve diagnostic precision, treatment efficacy, and long-term survival in pediatric cancer and other childhood catastrophic diseases. We present St. Jude Cloud (https://www.stjude.cloud), a cloud-based data-sharing ecosystem for accessing, analyzing, and visualizing genomic data from >10,000 pediatric patients with cancer and long-term survivors, and >800 pediatric sickle cell patients. Harmonized genomic data totaling 1.25 petabytes are freely available, including 12,104 whole genomes, 7,697 whole exomes, and 2,202 transcriptomes. The resource is expanding rapidly, with regular data uploads from St. Jude's prospective clinical genomics programs. Three interconnected apps within the ecosystem-Genomics Platform, Pediatric Cancer Knowledgebase, and Visualization Community-enable simultaneously performing advanced data analysis in the cloud and enhancing the Pediatric Cancer knowledgebase. We demonstrate the value of the ecosystem through use cases that classify 135 pediatric cancer subtypes by gene expression profiling and map mutational signatures across 35 pediatric cancer subtypes. SIGNIFICANCE: To advance research and treatment of pediatric cancer, we developed St. Jude Cloud, a data-sharing ecosystem for accessing >1.2 petabytes of raw genomic data from >10,000 pediatric patients and survivors, innovative analysis workflows, integrative multiomics visualizations, and a knowledgebase of published data contributed by the global pediatric cancer community.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Anemia, Sickle Cell/genetics , Cloud Computing , Genomics , Information Dissemination , Neoplasms/genetics , Child , Ecosystem , Hospitals, Pediatric , Humans
15.
Cancer Cell ; 39(1): 83-95.e4, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33434514

ABSTRACT

GenomePaint (https://genomepaint.stjude.cloud/) is an interactive visualization platform for whole-genome, whole-exome, transcriptome, and epigenomic data of tumor samples. Its design captures the inter-relatedness between DNA variations and RNA expression, supporting in-depth exploration of both individual cancer genomes and full cohorts. Regulatory non-coding variants can be inspected and analyzed along with coding variants, and their functional impact further explored by examining 3D genome data from cancer cell lines. Further, GenomePaint correlates mutation and expression patterns with patient outcomes, and supports custom data upload. We used GenomePaint to unveil aberrant splicing that disrupts the RING domain of CREBBP, discover cis activation of the MYC oncogene by duplication of the NOTCH1-MYC enhancer in B-lineage acute lymphoblastic leukemia, and explore the inter- and intra-tumor heterogeneity at EGFR in adult glioblastomas. These examples demonstrate that deep multi-omics exploration of individual cancer genomes enabled by GenomePaint can lead to biological insights for follow-up validation.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Genetic Variation , Neoplasms/genetics , Adult , Cell Line, Tumor , Child , Databases, Genetic , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Male , User-Computer Interface , Exome Sequencing , Whole Genome Sequencing
16.
Nat Genet ; 52(8): 811-818, 2020 08.
Article in English | MEDLINE | ID: mdl-32632335

ABSTRACT

We developed cis-X, a computational method for discovering regulatory noncoding variants in cancer by integrating whole-genome and transcriptome sequencing data from a single cancer sample. cis-X first finds aberrantly cis-activated genes that exhibit allele-specific expression accompanied by an elevated outlier expression. It then searches for causal noncoding variants that may introduce aberrant transcription factor binding motifs or enhancer hijacking by structural variations. Analysis of 13 T-lineage acute lymphoblastic leukemias identified a recurrent intronic variant predicted to cis-activate the TAL1 oncogene, a finding validated in vivo by chromatin immunoprecipitation sequencing of a patient-derived xenograft. Candidate oncogenes include the prolactin receptor PRLR activated by a focal deletion that removes a CTCF-insulated neighborhood boundary. cis-X may be applied to pediatric and adult solid tumors that are aneuploid and heterogeneous. In contrast to existing approaches, which require large sample cohorts, cis-X enables the discovery of regulatory noncoding variants in individual cancer genomes.


Subject(s)
Enhancer Elements, Genetic/genetics , Genetic Variation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Untranslated/genetics , Adolescent , Alleles , Child , Child, Preschool , Chromatin/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Oncogenes/genetics , Transcription, Genetic/genetics
17.
Genome Biol ; 21(1): 126, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32466770

ABSTRACT

To discover driver fusions beyond canonical exon-to-exon chimeric transcripts, we develop CICERO, a local assembly-based algorithm that integrates RNA-seq read support with extensive annotation for candidate ranking. CICERO outperforms commonly used methods, achieving a 95% detection rate for 184 independently validated driver fusions including internal tandem duplications and other non-canonical events in 170 pediatric cancer transcriptomes. Re-analysis of TCGA glioblastoma RNA-seq unveils previously unreported kinase fusions (KLHL7-BRAF) and a 13% prevalence of EGFR C-terminal truncation. Accessible via standard or cloud-based implementation, CICERO enhances driver fusion detection for research and precision oncology. The CICERO source code is available at https://github.com/stjude/Cicero.


Subject(s)
Gene Fusion , Molecular Sequence Annotation/methods , Neoplasms/genetics , Software , Algorithms , Humans , Sequence Analysis, RNA
18.
Blood ; 135(1): 41-55, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31697823

ABSTRACT

To study the mechanisms of relapse in acute lymphoblastic leukemia (ALL), we performed whole-genome sequencing of 103 diagnosis-relapse-germline trios and ultra-deep sequencing of 208 serial samples in 16 patients. Relapse-specific somatic alterations were enriched in 12 genes (NR3C1, NR3C2, TP53, NT5C2, FPGS, CREBBP, MSH2, MSH6, PMS2, WHSC1, PRPS1, and PRPS2) involved in drug response. Their prevalence was 17% in very early relapse (<9 months from diagnosis), 65% in early relapse (9-36 months), and 32% in late relapse (>36 months) groups. Convergent evolution, in which multiple subclones harbor mutations in the same drug resistance gene, was observed in 6 relapses and confirmed by single-cell sequencing in 1 case. Mathematical modeling and mutational signature analysis indicated that early relapse resistance acquisition was frequently a 2-step process in which a persistent clone survived initial therapy and later acquired bona fide resistance mutations during therapy. In contrast, very early relapses arose from preexisting resistant clone(s). Two novel relapse-specific mutational signatures, one of which was caused by thiopurine treatment based on in vitro drug exposure experiments, were identified in early and late relapses but were absent from 2540 pan-cancer diagnosis samples and 129 non-ALL relapses. The novel signatures were detected in 27% of relapsed ALLs and were responsible for 46% of acquired resistance mutations in NT5C2, PRPS1, NR3C1, and TP53. These results suggest that chemotherapy-induced drug resistance mutations facilitate a subset of pediatric ALL relapses.


Subject(s)
Biomarkers, Tumor/genetics , Methotrexate/therapeutic use , Mutagenesis/drug effects , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , 5'-Nucleotidase/genetics , Antimetabolites, Antineoplastic/therapeutic use , Child , DNA Mutational Analysis , Female , Follow-Up Studies , Genomics , High-Throughput Nucleotide Sequencing , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prognosis , Receptors, Glucocorticoid/genetics , Survival Rate , Tumor Suppressor Protein p53/genetics
19.
J Ind Microbiol Biotechnol ; 47(2): 209-222, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31853777

ABSTRACT

A highly efficient lycopene production system was constructed by assembling enzymes fused to zinc-finger motifs on DNA scaffolds in vitro and in vivo. Three key enzymes of the lycopene synthesis pathway, geranylgeranyl diphosphate synthase, phytoene synthase, and phytoene desaturase, were fused with zinc-finger proteins, expressed and purified. Recombinant plasmids of the pS series containing DNA scaffolds that the zinc-finger proteins can specifically bind to were constructed. In the in vitro system, the production efficiency of lycopene was improved greatly after the addition of the scaffold plasmid pS231. Subsequently, the plasmid pET-AEBI was constructed and introduced into recombinant Escherichia coli BL21 (DE3) for expression, together with plasmids of the pS series. The lycopene production rate and content of the recombinant strain pp231 were higher than that of all strains carrying the DNA scaffold and the control. With the addition of cofactors and substrates in the lycopene biosynthesis pathway, the lycopene yield of pp231 reached 632.49 mg/L at 40 h, representing a 4.7-fold increase compared to the original recombinant strain pA1A3. This DNA scaffold system can be used as a platform for the construction and production of many biochemicals synthesized via multi-enzyme cascade reactions.


Subject(s)
DNA/genetics , Lycopene/metabolism , DNA/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Oxidoreductases/metabolism , Plasmids/genetics , Zinc Fingers
20.
Nat Commun ; 10(1): 2789, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31243274

ABSTRACT

IGH@ proto-oncogene translocation is a common oncogenic event in lymphoid lineage cancers such as B-ALL, lymphoma and multiple myeloma. Here, to investigate the interplay between IGH@ proto-oncogene translocation and IGH allelic exclusion, we perform long-read whole-genome and transcriptome sequencing along with epigenetic and 3D genome profiling of Nalm6, an IGH-DUX4 positive B-ALL cell line. We detect significant allelic imbalance on the wild-type over the IGH-DUX4 haplotype in expression and epigenetic data, showing IGH-DUX4 translocation occurs on the silenced IGH allele. In vitro, this reduces the oncogenic stress of DUX4 high-level expression. Moreover, patient samples of IGH-DUX4 B-ALL have similar expression profile and IGH breakpoints as Nalm6, suggesting a common mechanism to allow optimal dosage of non-toxic DUX4 expression.


Subject(s)
Homeodomain Proteins/metabolism , Immunoglobulin Heavy Chains/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Apoptosis , DNA/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Genomics , Histones , Humans , Mice , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Mas , Whole Genome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...