Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Immunity ; 55(3): 512-526.e9, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263569

ABSTRACT

Dual blockade of the PD-1 and TIGIT coinhibitory receptors on T cells shows promising early results in cancer patients. Here, we studied the mechanisms whereby PD-1 and/or TIGIT blockade modulate anti-tumor CD8+ T cells. Although PD-1 and TIGIT are thought to regulate different costimulatory receptors (CD28 and CD226), effectiveness of PD-1 or TIGIT inhibition in preclinical tumor models was reduced in the absence of CD226. CD226 expression associated with clinical benefit in patients with non-small cell lung carcinoma (NSCLC) treated with anti-PD-L1 antibody atezolizumab. CD226 and CD28 were co-expressed on NSCLC infiltrating CD8+ T cells poised for expansion. Mechanistically, PD-1 inhibited phosphorylation of both CD226 and CD28 via its ITIM-containing intracellular domain (ICD); TIGIT's ICD was dispensable, with TIGIT restricting CD226 co-stimulation by blocking interaction with their common ligand PVR (CD155). Thus, full restoration of CD226 signaling, and optimal anti-tumor CD8+ T cell responses, requires blockade of TIGIT and PD-1, providing a mechanistic rationale for combinatorial targeting in the clinic.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Antigens, Differentiation, T-Lymphocyte/metabolism , CD28 Antigens/metabolism , Humans , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism
2.
Nat Commun ; 12(1): 7310, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911951

ABSTRACT

Inositol requiring enzyme 1 (IRE1) mitigates endoplasmic-reticulum (ER) stress by orchestrating the unfolded-protein response (UPR). IRE1 spans the ER membrane, and signals through a cytosolic kinase-endoribonuclease module. The endoribonuclease generates the transcription factor XBP1s by intron excision between similar RNA stem-loop endomotifs, and depletes select cellular mRNAs through regulated IRE1-dependent decay (RIDD). Paradoxically, in mammals RIDD seems to target only mRNAs with XBP1-like endomotifs, while in flies RIDD exhibits little sequence restriction. By comparing nascent and total IRE1α-controlled mRNAs in human cells, we identify not only canonical endomotif-containing RIDD substrates, but also targets without such motifs-degraded by a process we coin RIDDLE, for RIDD lacking endomotif. IRE1α displays two basic endoribonuclease modalities: highly specific, endomotif-directed cleavage, minimally requiring dimers; and more promiscuous, endomotif-independent processing, requiring phospho-oligomers. An oligomer-deficient IRE1α mutant fails to support RIDDLE in vitro and in cells. Our results advance current mechanistic understanding of the UPR.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Endoplasmic Reticulum/genetics , Endoribonucleases/genetics , Humans , Protein Serine-Threonine Kinases/genetics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Unfolded Protein Response
3.
J Immunother Cancer ; 9(10)2021 10.
Article in English | MEDLINE | ID: mdl-34599029

ABSTRACT

BACKGROUND: Individualized neoantigen-specific immunotherapy (iNeST) requires robustly expressed clonal neoantigens for efficacy, but tumor mutational heterogeneity, loss of neoantigen expression, and variable tissue sampling present challenges. It is assumed that clonal neoantigens are preferred targets for immunotherapy, but the distributions of clonal neoantigens are not well characterized across cancer types. METHODS: We combined multiregion sequencing (MR-seq) analysis of five untreated, synchronously sampled metastatic solid tumors with re-analysis of published MR-seq data from 103 patients in order to characterize their globally clonal neoantigen content and factors that would impact neoantigen targeting. RESULTS: Branching evolution in colorectal cancer and renal cell carcinoma led to fewer clonal neoantigens and to clade-specific neoantigens (those shared across a subset of tumor regions but not fully clonal), with the latter not being readily distinguishable in single tumor samples. In colorectal, renal, and bladder cancer, most tumors had few globally clonal neoantigens. Prioritizing mutations with higher purity-adjusted and ploidy-adjusted variant allele frequency enriched for globally clonal neoantigens (those found in all tumor regions), whereas estimated cancer cell fraction derived from clustering-based tools, surprisingly, did not. Neoantigen quality was associated with loss of neoantigen expression in the bladder cancer case, and HLA-allele loss was observed in the renal and non-small cell lung cancer cases. CONCLUSIONS: We show that tumor type, multilesion sampling, neoantigen expression, and HLA allele retention are important factors for iNeST targeting and patient selection, and may also be important factors to consider in the development of biomarker strategies.


Subject(s)
Antigens, Neoplasm/immunology , Biomarkers, Tumor/metabolism , Immunotherapy/methods , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Female , Humans , Male , Mice , Middle Aged , Neoplasms/genetics
4.
J Immunother Cancer ; 9(4)2021 04.
Article in English | MEDLINE | ID: mdl-33827905

ABSTRACT

BACKGROUND: CD8+ tissue-resident memory T (TRM) cells, marked by CD103 (ITGAE) expression, are thought to actively suppress cancer progression, leading to the hypothesis that their presence in tumors may predict response to immunotherapy. METHODS: Here, we test this by combining high-dimensional single-cell modalities with bulk tumor transcriptomics from 1868 patients enrolled in lung and bladder cancer clinical trials of atezolizumab (anti-programmed cell death ligand 1 (PD-L1)). RESULTS: ITGAE was identified as the most significantly upregulated gene in inflamed tumors. Tumor CD103+ CD8+ TRM cells exhibited a complex phenotype defined by the expression of checkpoint regulators, cytotoxic proteins, and increased clonal expansion. CONCLUSIONS: Our analyses indeed demonstrate that the presence of CD103+ CD8+ TRM cells, quantified by tracking intratumoral CD103 expression, can predict treatment outcome, suggesting that patients who respond to PD-1/PD-L1 blockade are those who exhibit an ongoing antitumor T-cell response.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, CD/genetics , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , Immune Checkpoint Inhibitors/therapeutic use , Integrin alpha Chains/genetics , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/immunology , Urinary Bladder Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/adverse effects , B7-H1 Antigen/immunology , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Databases, Genetic , Gene Expression Profiling , Humans , Immune Checkpoint Inhibitors/adverse effects , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Phenotype , Randomized Controlled Trials as Topic , Time Factors , Treatment Outcome , Tumor Microenvironment , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
5.
Cancer Res ; 80(11): 2368-2379, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32265225

ABSTRACT

Cancer cells exploit the unfolded protein response (UPR) to mitigate endoplasmic reticulum (ER) stress caused by cellular oncogene activation and a hostile tumor microenvironment (TME). The key UPR sensor IRE1α resides in the ER and deploys a cytoplasmic kinase-endoribonuclease module to activate the transcription factor XBP1s, which facilitates ER-mediated protein folding. Studies of triple-negative breast cancer (TNBC)-a highly aggressive malignancy with a dismal posttreatment prognosis-implicate XBP1s in promoting tumor vascularization and progression. However, it remains unknown whether IRE1α adapts the ER in TNBC cells and modulates their TME, and whether IRE1α inhibition can enhance antiangiogenic therapy-previously found to be ineffective in patients with TNBC. To gauge IRE1α function, we defined an XBP1s-dependent gene signature, which revealed significant IRE1α pathway activation in multiple solid cancers, including TNBC. IRE1α knockout in TNBC cells markedly reversed substantial ultrastructural expansion of their ER upon growth in vivo. IRE1α disruption also led to significant remodeling of the cellular TME, increasing pericyte numbers while decreasing cancer-associated fibroblasts and myeloid-derived suppressor cells. Pharmacologic IRE1α kinase inhibition strongly attenuated growth of cell line-based and patient-derived TNBC xenografts in mice and synergized with anti-VEGFA treatment to cause tumor stasis or regression. Thus, TNBC cells critically rely on IRE1α to adapt their ER to in vivo stress and to adjust the TME to facilitate malignant growth. TNBC reliance on IRE1α is an important vulnerability that can be uniquely exploited in combination with antiangiogenic therapy as a promising new biologic approach to combat this lethal disease. SIGNIFICANCE: Pharmacologic IRE1α kinase inhibition reverses ultrastructural distension of the ER, normalizes the tumor vasculature, and remodels the cellular TME, attenuating TNBC growth in mice.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/therapy , Animals , Antineoplastic Agents, Immunological/immunology , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Female , Gene Knockout Techniques , Humans , Mice , Mice, SCID , Neovascularization, Pathologic/therapy , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Triple Negative Breast Neoplasms/blood supply , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , X-Box Binding Protein 1/antagonists & inhibitors , X-Box Binding Protein 1/genetics , Xenograft Model Antitumor Assays
6.
Nature ; 579(7798): 274-278, 2020 03.
Article in English | MEDLINE | ID: mdl-32103181

ABSTRACT

Despite the resounding clinical success in cancer treatment of antibodies that block the interaction of PD1 with its ligand PDL11, the mechanisms involved remain unknown. A major limitation to understanding the origin and fate of T cells in tumour immunity is the lack of quantitative information on the distribution of individual clonotypes of T cells in patients with cancer. Here, by performing deep single-cell sequencing of RNA and T cell receptors in patients with different types of cancer, we survey the profiles of various populations of T cells and T cell receptors in tumours, normal adjacent tissue, and peripheral blood. We find clear evidence of clonotypic expansion of effector-like T cells not only within the tumour but also in normal adjacent tissue. Patients with gene signatures of such clonotypic expansion respond best to anti-PDL1 therapy. Notably, expanded clonotypes found in the tumour and normal adjacent tissue can also typically be detected in peripheral blood, which suggests a convenient approach to patient identification. Analyses of our data together with several external datasets suggest that intratumoural T cells, especially in responsive patients, are replenished with fresh, non-exhausted replacement cells from sites outside the tumour, suggesting continued activity of the cancer immunity cycle in these patients, the acceleration of which may be associated with clinical response.


Subject(s)
Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/pathology , Pharmacogenomic Variants , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Clone Cells , Humans , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes/metabolism , Transcriptome
7.
Proc Natl Acad Sci U S A ; 115(50): E11731-E11740, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30504141

ABSTRACT

Natural killer (NK) cell recognition of tumor cells is mediated through activating receptors such as CD226, with suppression of effector functions often controlled by negative regulatory transcription factors such as FOXO1. Here we show that CD226 regulation of NK cell cytotoxicity is facilitated through inactivation of FOXO1. Gene-expression analysis of NK cells isolated from syngeneic tumors grown in wild-type or CD226-deficient mice revealed dysregulated expression of FOXO1-regulated genes in the absence of CD226. In vitro cytotoxicity and stimulation assays demonstrated that CD226 is required for optimal killing of tumor target cells, with engagement of its ligand CD155 resulting in phosphorylation of FOXO1. CD226 deficiency or anti-CD226 antibody blockade impaired cytotoxicity with concomitant compromised inactivation of FOXO1. Furthermore, inhibitors of FOXO1 phosphorylation abrogated CD226-mediated signaling and effector responses. These results define a pathway by which CD226 exerts control of NK cell responses against tumors.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic , Gene Expression Regulation, Neoplastic , Humans , Ligands , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Knockout , Nectins/metabolism , Phosphorylation , Receptors, Virus/metabolism , Signal Transduction/immunology
8.
Cancer Cell ; 31(3): 383-395, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28262555

ABSTRACT

The anti-FcRH5/CD3 T cell-dependent bispecific antibody (TDB) targets the B cell lineage marker FcRH5 expressed in multiple myeloma (MM) tumor cells. We demonstrate that TDBs trigger T cell receptor activation by inducing target clustering and exclusion of CD45 phosphatase from the synapse. The dimensions of the target molecule play a key role in the efficiency of the synapse formation. The anti-FcRH5/CD3 TDB kills human plasma cells and patient-derived myeloma cells at picomolar concentrations and results in complete depletion of B cells and bone marrow plasma cells in cynomolgus monkeys. These data demonstrate the potential for the anti-FcRH5/CD3 TDB, alone or in combination with inhibition of PD-1/PD-L1 signaling, in the treatment of MM and other B cell malignancies.


Subject(s)
Antibodies, Bispecific/therapeutic use , CD3 Complex/immunology , Epitopes , Immunological Synapses/physiology , Multiple Myeloma/drug therapy , Receptors, Fc/immunology , T-Lymphocytes/immunology , Animals , Cytokines/metabolism , Humans , Leukocyte Common Antigens/physiology , Lymphocyte Activation , Macaca fascicularis , Mice , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Programmed Cell Death 1 Receptor/physiology , Receptors, Antigen, T-Cell/physiology , Receptors, Fc/analysis
9.
PLoS Pathog ; 12(6): e1005702, 2016 06.
Article in English | MEDLINE | ID: mdl-27351973

ABSTRACT

Broadly neutralizing antibodies targeting the stalk region of influenza A virus (IAV) hemagglutinin (HA) are effective in blocking virus infection both in vitro and in vivo. The highly conserved epitopes recognized by these antibodies are critical for the membrane fusion function of HA and therefore less likely to be permissive for virus mutational escape. Here we report three resistant viruses of the A/Perth/16/2009 strain that were selected in the presence of a broadly neutralizing stalk-binding antibody. The three resistant viruses harbor three different mutations in the HA stalk: (1) Gln387Lys; (2) Asp391Tyr; (3) Asp391Gly. The Gln387Lys mutation completely abolishes binding of the antibody to the HA stalk epitope. The other two mutations, Asp391Tyr and Asp391Gly, do not affect antibody binding at neutral pH and only slightly reduce binding at low pH. Interestingly, they enhance the fusion ability of the HA, representing a novel mechanism that allows productive membrane fusion even in the presence of antibody and hence virus escape from antibody neutralization. Therefore, these mutations illustrate two different resistance mechanisms used by IAV to escape broadly neutralizing stalk-binding antibodies. Compared to the wild type virus, the resistant viruses release fewer progeny viral particles during replication and are more sensitive to Tamiflu, suggesting reduced viral fitness.


Subject(s)
Antibodies, Neutralizing/immunology , Drug Resistance, Microbial/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immune Evasion/immunology , Influenza A virus/immunology , Animals , Antibodies, Viral/immunology , Blotting, Western , Dogs , Flow Cytometry , Humans , Immunohistochemistry , Influenza, Human/immunology , Madin Darby Canine Kidney Cells , Mice , Neutralization Tests , Orthomyxoviridae Infections/immunology , Polymerase Chain Reaction
10.
PLoS One ; 11(5): e0156132, 2016.
Article in English | MEDLINE | ID: mdl-27218464

ABSTRACT

Analysis of splice variants from short read RNA-seq data remains a challenging problem. Here we present a novel method for the genome-guided prediction and quantification of splice events from RNA-seq data, which enables the analysis of unannotated and complex splice events. Splice junctions and exons are predicted from reads mapped to a reference genome and are assembled into a genome-wide splice graph. Splice events are identified recursively from the graph and are quantified locally based on reads extending across the start or end of each splice variant. We assess prediction accuracy based on simulated and real RNA-seq data, and illustrate how different read aligners (GSNAP, HISAT2, STAR, TopHat2) affect prediction results. We validate our approach for quantification based on simulated data, and compare local estimates of relative splice variant usage with those from other methods (MISO, Cufflinks) based on simulated and real RNA-seq data. In a proof-of-concept study of splice variants in 16 normal human tissues (Illumina Body Map 2.0) we identify 249 internal exons that belong to known genes but are not related to annotated exons. Using independent RNA samples from 14 matched normal human tissues, we validate 9/9 of these exons by RT-PCR and 216/249 by paired-end RNA-seq (2 x 250 bp). These results indicate that de novo prediction of splice variants remains beneficial even in well-studied systems. An implementation of our method is freely available as an R/Bioconductor package [Formula: see text].


Subject(s)
Computational Biology/methods , RNA Splicing , RNA/genetics , Sequence Analysis, RNA/methods , Algorithms , Alternative Splicing , Exons , Humans , Software
11.
Algorithms Mol Biol ; 11: 5, 2016.
Article in English | MEDLINE | ID: mdl-27095998

ABSTRACT

BACKGROUND: Hash tables constitute a widely used data structure for indexing genomes that provides a list of genomic positions for each possible oligomer of a given size. The offset array in a hash table grows exponentially with the oligomer size and precludes the use of larger oligomers that could facilitate rapid alignment of sequences to a genome. RESULTS: We propose to compress the offset array using vectorized bitpacking. We introduce an algorithm and data structure called BP64-columnar that achieves fast random access in arrays of monotonically nondecreasing integers. Experimental results based on hash tables for the fly, chicken, and human genomes show that BP64-columnar is 3 to 4 times faster than publicly available implementations of universal coding schemes, such as Elias gamma, Elias delta, and Fibonacci compression. Furthermore, among vectorized bitpacking schemes, our BP64-columnar format yields retrieval times that are faster than the fastest known bitpacking format by a factor of 3 for retrieving a single value, and a factor of 2 for retrieving two adjacent values. CONCLUSIONS: Our BP64-columnar scheme enables compression of genomic hash tables with fast retrieval. It also has potential applications to other domains requiring differential coding with random access.

12.
Algorithms Mol Biol ; 11: 9, 2016.
Article in English | MEDLINE | ID: mdl-27110277

ABSTRACT

BACKGROUND: Suffix arrays and their variants are used widely for representing genomes in search applications. Enhanced suffix arrays (ESAs) provide fast search speed, but require large auxiliary data structures for storing longest common prefix and child interval information. We explore techniques for compressing ESAs to accelerate genomic search and reduce memory requirements. RESULTS: We evaluate various bitpacking techniques that store integers in fewer than 32 bits each, as well as bytecoding methods that reserve a single byte per integer whenever possible. Our results on the fly, chicken, and human genomes show that bytecoding with an exception guide array is the fastest method for retrieving auxiliary information. Genomic searching can be further accelerated using a data structure called a discriminating character array, which reduces memory accesses to the suffix array and the genome string. Finally, integrating storage of the auxiliary and discriminating character arrays further speeds up genomic search. CONCLUSIONS: The combination of exception guide arrays, a discriminating character array, and integrated data storage provide a 2- to 3-fold increase in speed for genomic searching compared with using bytecoding alone, and is 20 % faster and 40 % more space-efficient than an uncompressed ESA.

13.
Methods Mol Biol ; 1418: 283-334, 2016.
Article in English | MEDLINE | ID: mdl-27008021

ABSTRACT

The programs GMAP and GSNAP, for aligning RNA-Seq and DNA-Seq datasets to genomes, have evolved along with advances in biological methodology to handle longer reads, larger volumes of data, and new types of biological assays. The genomic representation has been improved to include linear genomes that can compare sequences using single-instruction multiple-data (SIMD) instructions, compressed genomic hash tables with fast access using SIMD instructions, handling of large genomes with more than four billion bp, and enhanced suffix arrays (ESAs) with novel data structures for fast access. Improvements to the algorithms have included a greedy match-and-extend algorithm using suffix arrays, segment chaining using genomic hash tables, diagonalization using segmental hash tables, and nucleotide-level dynamic programming procedures that use SIMD instructions and eliminate the need for F-loop calculations. Enhancements to the functionality of the programs include standardization of indel positions, handling of ambiguous splicing, clipping and merging of overlapping paired-end reads, and alignments to circular chromosomes and alternate scaffolds. The programs have been adapted for use in pipelines by integrating their usage into R/Bioconductor packages such as gmapR and HTSeqGenie, and these pipelines have facilitated the discovery of numerous biological phenomena.


Subject(s)
Computational Biology/methods , Genomics/methods , Sequence Alignment/methods , Sequence Analysis, DNA/methods , Software , Algorithms , Databases, Genetic , Gene Expression Profiling/methods , Genome , Genomics/standards , High-Throughput Nucleotide Sequencing , Polymorphism, Single Nucleotide , RNA Editing , RNA Splicing , Reproducibility of Results , Sequence Analysis, DNA/standards , Time Factors
14.
Nat Genet ; 48(4): 407-16, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26928227

ABSTRACT

We analyzed transcriptomes (n = 211), whole exomes (n = 99) and targeted exomes (n = 103) from 216 malignant pleural mesothelioma (MPM) tumors. Using RNA-seq data, we identified four distinct molecular subtypes: sarcomatoid, epithelioid, biphasic-epithelioid (biphasic-E) and biphasic-sarcomatoid (biphasic-S). Through exome analysis, we found BAP1, NF2, TP53, SETD2, DDX3X, ULK2, RYR2, CFAP45, SETDB1 and DDX51 to be significantly mutated (q-score ≥ 0.8) in MPMs. We identified recurrent mutations in several genes, including SF3B1 (∼2%; 4/216) and TRAF7 (∼2%; 5/216). SF3B1-mutant samples showed a splicing profile distinct from that of wild-type tumors. TRAF7 alterations occurred primarily in the WD40 domain and were, except in one case, mutually exclusive with NF2 alterations. We found recurrent gene fusions and splice alterations to be frequent mechanisms for inactivation of NF2, BAP1 and SETD2. Through integrated analyses, we identified alterations in Hippo, mTOR, histone methylation, RNA helicase and p53 signaling pathways in MPMs.


Subject(s)
Lung Neoplasms/genetics , Mesothelioma/genetics , Oncogene Proteins, Fusion/genetics , Pleural Neoplasms/genetics , Alternative Splicing , Cell Line, Tumor , DNA Mutational Analysis , Exome , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Mesothelioma/metabolism , Mesothelioma/mortality , Mesothelioma, Malignant , Mutation , Oncogene Proteins, Fusion/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pleural Neoplasms/metabolism , Pleural Neoplasms/mortality , Polymorphism, Single Nucleotide , Proportional Hazards Models , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing Factors , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoprotein, U2 Small Nuclear/metabolism
15.
Nat Genet ; 47(1): 13-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25401301

ABSTRACT

To further understand the molecular distinctions between kidney cancer subtypes, we analyzed exome, transcriptome and copy number alteration data from 167 primary human tumors that included renal oncocytomas and non-clear cell renal cell carcinomas (nccRCCs), consisting of papillary (pRCC), chromophobe (chRCC) and translocation (tRCC) subtypes. We identified ten significantly mutated genes in pRCC, including MET, NF2, SLC5A3, PNKD and CPQ. MET mutations occurred in 15% (10/65) of pRCC samples and included previously unreported recurrent activating mutations. In chRCC, we found TP53, PTEN, FAAH2, PDHB, PDXDC1 and ZNF765 to be significantly mutated. Gene expression analysis identified a five-gene set that enabled the molecular classification of chRCC, renal oncocytoma and pRCC. Using RNA sequencing, we identified previously unreported gene fusions, including ACTG1-MITF fusion. Ectopic expression of the ACTG1-MITF fusion led to cellular transformation and induced the expression of downstream target genes. Finally, we observed upregulation of the anti-apoptotic factor BIRC7 in MiTF-high RCC tumors, suggesting a potential therapeutic role for BIRC7 inhibitors.


Subject(s)
Carcinoma, Renal Cell/classification , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Mutation , Adenoma, Oxyphilic/classification , Adenoma, Oxyphilic/genetics , Adenoma, Oxyphilic/pathology , Amino Acid Sequence , Base Sequence , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , DNA, Neoplasm , Gene Dosage , Genomic Instability , Humans , Kidney Neoplasms/classification , Kidney Neoplasms/pathology , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Polymorphism, Single Nucleotide , Protein Conformation , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , Translocation, Genetic
16.
Nat Biotechnol ; 33(3): 306-12, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25485619

ABSTRACT

Tumor-derived cell lines have served as vital models to advance our understanding of oncogene function and therapeutic responses. Although substantial effort has been made to define the genomic constitution of cancer cell line panels, the transcriptome remains understudied. Here we describe RNA sequencing and single-nucleotide polymorphism (SNP) array analysis of 675 human cancer cell lines. We report comprehensive analyses of transcriptome features including gene expression, mutations, gene fusions and expression of non-human sequences. Of the 2,200 gene fusions catalogued, 1,435 consist of genes not previously found in fusions, providing many leads for further investigation. We combine multiple genome and transcriptome features in a pathway-based approach to enhance prediction of response to targeted therapeutics. Our results provide a valuable resource for studies that use cancer cell lines.


Subject(s)
Neoplasms/genetics , Transcription, Genetic , Base Sequence , Cell Line, Tumor , Cluster Analysis , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Mutation/genetics , Oncogene Fusion/genetics , Organ Specificity/genetics , Polymorphism, Single Nucleotide/genetics
17.
Genome Biol ; 15(8): 436, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25159915

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a heterogeneous disease with high mortality rate. Recent genomic studies have identified TP53, AXIN1, and CTNNB1 as the most frequently mutated genes. Lower frequency mutations have been reported in ARID1A, ARID2 and JAK1. In addition, hepatitis B virus (HBV) integrations into the human genome have been associated with HCC. RESULTS: Here, we deep-sequence 42 HCC patients with a combination of whole genome, exome and transcriptome sequencing to identify the mutational landscape of HCC using a reasonably large discovery cohort. We find frequent mutations in TP53, CTNNB1 and AXIN1, and rare but likely functional mutations in BAP1 and IDH1. Besides frequent hepatitis B virus integrations at TERT, we identify translocations at the boundaries of TERT. A novel deletion is identified in CTNNB1 in a region that is heavily mutated in multiple cancers. We also find multiple high-allelic frequency mutations in the extracellular matrix protein LAMA2. Lower expression levels of LAMA2 correlate with a proliferative signature, and predict poor survival and higher chance of cancer recurrence in HCC patients, suggesting an important role of the extracellular matrix and cell adhesion in tumor progression of a subgroup of HCC patients. CONCLUSIONS: The heterogeneous disease of HCC features diverse modes of genomic alteration. In addition to common point mutations, structural variations and methylation changes, there are several virus-associated changes, including gene disruption or activation, formation of chimeric viral-human transcripts, and DNA copy number changes. Such a multitude of genomic events likely contributes to the heterogeneous nature of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Mutational Analysis/methods , Genetic Variation , Laminin/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/virology , Genetic Heterogeneity , Hepatitis B/genetics , Hepatitis B virus/physiology , High-Throughput Nucleotide Sequencing , Humans , Liver Neoplasms/virology , Mutation Rate , Survival Analysis
18.
Nat Commun ; 5: 3830, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24807215

ABSTRACT

Gastric cancer is the second leading cause of worldwide cancer mortality, yet the underlying genomic alterations remain poorly understood. Here we perform exome and transcriptome sequencing and SNP array assays to characterize 51 primary gastric tumours and 32 cell lines. Meta-analysis of exome data and previously published data sets reveals 24 significantly mutated genes in microsatellite stable (MSS) tumours and 16 in microsatellite instable (MSI) tumours. Over half the patients in our collection could potentially benefit from targeted therapies. We identify 55 splice site mutations accompanied by aberrant splicing products, in addition to mutation-independent differential isoform usage in tumours. ZAK kinase isoform TV1 is preferentially upregulated in gastric tumours and cell lines relative to normal samples. This pattern is also observed in colorectal, bladder and breast cancers. Overexpression of this particular isoform activates multiple cancer-related transcription factor reporters, while depletion of ZAK in gastric cell lines inhibits proliferation. These results reveal the spectrum of genomic and transcriptomic alterations in gastric cancer, and identify isoform-specific oncogenic properties of ZAK.


Subject(s)
Protein Isoforms/genetics , Protein Kinases/genetics , Stomach Neoplasms/genetics , Base Sequence , Cell Line , Cell Proliferation/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , MAP Kinase Kinase Kinases , Microsatellite Instability , Microsatellite Repeats/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Receptor, ErbB-2/genetics , Sequence Analysis, DNA , Transcriptome/genetics
19.
Proc Natl Acad Sci U S A ; 109(47): 19368-73, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23134728

ABSTRACT

The protein kinase v-akt murine thymoma viral oncogene homolog (AKT), a key regulator of cell survival and proliferation, is frequently hyperactivated in human cancers. Intramolecular pleckstrin homology (PH) domain-kinase domain (KD) interactions are important in maintaining AKT in an inactive state. AKT activation proceeds after a conformational change that dislodges the PH from the KD. To understand these autoinhibitory interactions, we generated mutations at the PH-KD interface and found that most of them lead to constitutive activation of AKT. Such mutations are likely another mechanism by which activation may occur in human cancers and other diseases. In support of this likelihood, we found somatic mutations in AKT1 at the PH-KD interface that have not been previously described in human cancers. Furthermore, we show that the AKT1 somatic mutants are constitutively active, leading to oncogenic signaling. Additionally, our studies show that the AKT1 mutants are not effectively inhibited by allosteric AKT inhibitors, consistent with the requirement for an intact PH-KD interface for allosteric inhibition. These results have important implications for therapeutic intervention in patients with AKT mutations at the PH-KD interface.


Subject(s)
Neoplasms/enzymology , Neoplasms/genetics , Oncogenes/genetics , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Enzyme Activation/drug effects , Humans , Mice , Models, Molecular , Mutant Proteins/metabolism , Mutation/genetics , NIH 3T3 Cells , Protein Binding/drug effects , Protein Binding/genetics , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Genome Res ; 22(12): 2315-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23033341

ABSTRACT

Lung cancer is a highly heterogeneous disease in terms of both underlying genetic lesions and response to therapeutic treatments. We performed deep whole-genome sequencing and transcriptome sequencing on 19 lung cancer cell lines and three lung tumor/normal pairs. Overall, our data show that cell line models exhibit similar mutation spectra to human tumor samples. Smoker and never-smoker cancer samples exhibit distinguishable patterns of mutations. A number of epigenetic regulators, including KDM6A, ASH1L, SMARCA4, and ATAD2, are frequently altered by mutations or copy number changes. A systematic survey of splice-site mutations identified 106 splice site mutations associated with cancer specific aberrant splicing, including mutations in several known cancer-related genes. RAC1b, an isoform of the RAC1 GTPase that includes one additional exon, was found to be preferentially up-regulated in lung cancer. We further show that its expression is significantly associated with sensitivity to a MAP2K (MEK) inhibitor PD-0325901. Taken together, these data present a comprehensive genomic landscape of a large number of lung cancer samples and further demonstrate that cancer-specific alternative splicing is a widespread phenomenon that has potential utility as therapeutic biomarkers. The detailed characterizations of the lung cancer cell lines also provide genomic context to the vast amount of experimental data gathered for these lines over the decades, and represent highly valuable resources for cancer biology.


Subject(s)
Alternative Splicing , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Lung Neoplasms/genetics , Mutation , Transcriptome , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Line, Tumor , DNA Copy Number Variations , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenomics , Exons , Genetic Markers , Heterozygote , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase , Humans , Karyotyping/methods , Lung Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymorphism, Single Nucleotide , Reproducibility of Results , Sequence Analysis, RNA , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...