Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
Add more filters

Publication year range
1.
Cell ; 173(3): 611-623.e17, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29656891

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by near-universal loss of the short arm of chromosome 3, deleting several tumor suppressor genes. We analyzed whole genomes from 95 biopsies across 33 patients with clear cell renal cell carcinoma. We find hotspots of point mutations in the 5' UTR of TERT, targeting a MYC-MAX-MAD1 repressor associated with telomere lengthening. The most common structural abnormality generates simultaneous 3p loss and 5q gain (36% patients), typically through chromothripsis. This event occurs in childhood or adolescence, generally as the initiating event that precedes emergence of the tumor's most recent common ancestor by years to decades. Similar genomic changes drive inherited ccRCC. Modeling differences in age incidence between inherited and sporadic cancers suggests that the number of cells with 3p loss capable of initiating sporadic tumors is no more than a few hundred. Early development of ccRCC follows well-defined evolutionary trajectories, offering opportunity for early intervention.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Disease Progression , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mutation , 5' Untranslated Regions , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 5 , Female , Gene Dosage , Genome, Human , Humans , Male , Middle Aged , Prospective Studies , Telomerase/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
2.
Cell ; 173(3): 595-610.e11, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29656894

ABSTRACT

The evolutionary features of clear-cell renal cell carcinoma (ccRCC) have not been systematically studied to date. We analyzed 1,206 primary tumor regions from 101 patients recruited into the multi-center prospective study, TRACERx Renal. We observe up to 30 driver events per tumor and show that subclonal diversification is associated with known prognostic parameters. By resolving the patterns of driver event ordering, co-occurrence, and mutual exclusivity at clone level, we show the deterministic nature of clonal evolution. ccRCC can be grouped into seven evolutionary subtypes, ranging from tumors characterized by early fixation of multiple mutational and copy number drivers and rapid metastases to highly branched tumors with >10 subclonal drivers and extensive parallel evolution associated with attenuated progression. We identify genetic diversity and chromosomal complexity as determinants of patient outcome. Our insights reconcile the variable clinical behavior of ccRCC and suggest evolutionary potential as a biomarker for both intervention and surveillance.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Alleles , Biomarkers, Tumor , Chromosomes , Clonal Evolution , Disease Progression , Evolution, Molecular , Female , Genetic Heterogeneity , Genetic Variation , Humans , Longitudinal Studies , Male , Middle Aged , Models, Statistical , Mutation , Neoplasm Metastasis , Phenotype , Phylogeny , Prognosis , Prospective Studies , Sequence Analysis, DNA
3.
Cell ; 173(3): 581-594.e12, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29656895

ABSTRACT

Clear-cell renal cell carcinoma (ccRCC) exhibits a broad range of metastatic phenotypes that have not been systematically studied to date. Here, we analyzed 575 primary and 335 metastatic biopsies across 100 patients with metastatic ccRCC, including two cases sampledat post-mortem. Metastatic competence was afforded by chromosome complexity, and we identify 9p loss as a highly selected event driving metastasis and ccRCC-related mortality (p = 0.0014). Distinct patterns of metastatic dissemination were observed, including rapid progression to multiple tissue sites seeded by primary tumors of monoclonal structure. By contrast, we observed attenuated progression in cases characterized by high primary tumor heterogeneity, with metastatic competence acquired gradually and initial progression to solitary metastasis. Finally, we observed early divergence of primitive ancestral clones and protracted latency of up to two decades as a feature of pancreatic metastases.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mutation , Neoplasm Metastasis , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Biopsy , Chromosome Mapping , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 9 , Disease Progression , Female , Humans , Longitudinal Studies , Male , Middle Aged , Phenotype , Prospective Studies , Thrombosis , Treatment Outcome
4.
Mol Cell ; 81(4): 767-783.e11, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33333017

ABSTRACT

Chromatin is a barrier to efficient DNA repair, as it hinders access and processing of certain DNA lesions. ALC1/CHD1L is a nucleosome-remodeling enzyme that responds to DNA damage, but its precise function in DNA repair remains unknown. Here we report that loss of ALC1 confers sensitivity to PARP inhibitors, methyl-methanesulfonate, and uracil misincorporation, which reflects the need to remodel nucleosomes following base excision by DNA glycosylases but prior to handover to APEX1. Using CRISPR screens, we establish that ALC1 loss is synthetic lethal with homologous recombination deficiency (HRD), which we attribute to chromosome instability caused by unrepaired DNA gaps at replication forks. In the absence of ALC1 or APEX1, incomplete processing of BER intermediates results in post-replicative DNA gaps and a critical dependence on HR for repair. Hence, targeting ALC1 alone or as a PARP inhibitor sensitizer could be employed to augment existing therapeutic strategies for HRD cancers.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Nucleosomes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , DNA Helicases/genetics , DNA Replication/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/genetics , Homologous Recombination/drug effects , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Nucleosomes/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics
5.
Cell ; 153(5): 1050-63, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706742

ABSTRACT

RAS proteins are important direct activators of p110α, p110γ, and p110δ type I phosphoinositide 3-kinases (PI3Ks), interacting via an amino-terminal RAS-binding domain (RBD). Here, we investigate the regulation of the ubiquitous p110ß isoform of PI3K, implicated in G-protein-coupled receptor (GPCR) signaling, PTEN-loss-driven cancers, and thrombocyte function. Unexpectedly, RAS is unable to interact with p110ß, but instead RAC1 and CDC42 from the RHO subfamily of small GTPases bind and activate p110ß via its RBD. In fibroblasts, GPCRs couple to PI3K through Dock180/Elmo1-mediated RAC activation and subsequent interaction with p110ß. Cells from mice carrying mutations in the p110ß RBD show reduced PI3K activity and defective chemotaxis, and these mice are resistant to experimental lung fibrosis. These findings revise our understanding of the regulation of type I PI3K by showing that both RAS and RHO family GTPases directly regulate distinct ubiquitous PI3K isoforms and that RAC activates p110ß downstream of GPCRs.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Fibroblasts/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Chemotaxis , Class I Phosphatidylinositol 3-Kinases/chemistry , Fibrosis/chemically induced , Fibrosis/prevention & control , GTP-Binding Protein Regulators/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Isoenzymes/metabolism , Lung/pathology , Mice , Protein Interaction Domains and Motifs , rac1 GTP-Binding Protein/metabolism , ras Proteins/chemistry
6.
Cell ; 149(3): 642-55, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541434

ABSTRACT

Non-small cell lung cancer (NSCLC) is the most frequent cause of cancer deaths worldwide; nearly half contain mutations in the receptor tyrosine kinase/RAS pathway. Here we show that RAS-pathway mutant NSCLC cells depend on the transcription factor GATA2. Loss of GATA2 reduced the viability of NSCLC cells with RAS-pathway mutations, whereas wild-type cells were unaffected. Integrated gene expression and genome occupancy analyses revealed GATA2 regulation of the proteasome, and IL-1-signaling, and Rho-signaling pathways. These pathways were functionally significant, as reactivation rescued viability after GATA2 depletion. In a Kras-driven NSCLC mouse model, Gata2 loss dramatically reduced tumor development. Furthermore, Gata2 deletion in established Kras mutant tumors induced striking regression. Although GATA2 itself is likely undruggable, combined suppression of GATA2-regulated pathways with clinically approved inhibitors caused marked tumor clearance. Discovery of the nononcogene addiction of KRAS mutant lung cancers to GATA2 presents a network of druggable pathways for therapeutic exploitation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , GATA2 Transcription Factor/metabolism , Gene Regulatory Networks , Lung Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , ras Proteins/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , ras Proteins/genetics
7.
Mol Cell ; 70(4): 707-721.e7, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29754823

ABSTRACT

DNA polymerase ε (POLE) is a four-subunit complex and the major leading strand polymerase in eukaryotes. Budding yeast orthologs of POLE3 and POLE4 promote Polε processivity in vitro but are dispensable for viability in vivo. Here, we report that POLE4 deficiency in mice destabilizes the entire Polε complex, leading to embryonic lethality in inbred strains and extensive developmental abnormalities, leukopenia, and tumor predisposition in outbred strains. Comparable phenotypes of growth retardation and immunodeficiency are also observed in human patients harboring destabilizing mutations in POLE1. In both Pole4-/- mouse and POLE1 mutant human cells, Polε hypomorphy is associated with replication stress and p53 activation, which we attribute to inefficient replication origin firing. Strikingly, removing p53 is sufficient to rescue embryonic lethality and all developmental abnormalities in Pole4 null mice. However, Pole4-/-p53+/- mice exhibit accelerated tumorigenesis, revealing an important role for controlled CMG and origin activation in normal development and tumor prevention.


Subject(s)
Carcinogenesis/pathology , DNA Polymerase II/chemistry , DNA Polymerase II/physiology , DNA Replication , Developmental Disabilities/etiology , Growth Disorders/etiology , Leukopenia/etiology , Animals , Carcinogenesis/genetics , Cells, Cultured , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Humans , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Tumor Suppressor Protein p53/physiology
8.
Nature ; 514(7523): 498-502, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25341788

ABSTRACT

After immunogenic challenge, infiltrating and dividing lymphocytes markedly increase lymph node cellularity, leading to organ expansion. Here we report that the physical elasticity of lymph nodes is maintained in part by podoplanin (PDPN) signalling in stromal fibroblastic reticular cells (FRCs) and its modulation by CLEC-2 expressed on dendritic cells. We show in mouse cells that PDPN induces actomyosin contractility in FRCs via activation of RhoA/C and downstream Rho-associated protein kinase (ROCK). Engagement by CLEC-2 causes PDPN clustering and rapidly uncouples PDPN from RhoA/C activation, relaxing the actomyosin cytoskeleton and permitting FRC stretching. Notably, administration of CLEC-2 protein to immunized mice augments lymph node expansion. In contrast, lymph node expansion is significantly constrained in mice selectively lacking CLEC-2 expression in dendritic cells. Thus, the same dendritic cells that initiate immunity by presenting antigens to T lymphocytes also initiate remodelling of lymph nodes by delivering CLEC-2 to FRCs. CLEC-2 modulation of PDPN signalling permits FRC network stretching and allows for the rapid lymph node expansion--driven by lymphocyte influx and proliferation--that is the critical hallmark of adaptive immunity.


Subject(s)
Dendritic Cells/physiology , Fibroblasts/cytology , Lymph Nodes/cytology , Stromal Cells/cytology , Actomyosin/metabolism , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Dendritic Cells/immunology , Female , Fibroblasts/physiology , Inflammation/immunology , Lectins, C-Type/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Stromal Cells/physiology , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein , rhoC GTP-Binding Protein
9.
Nature ; 502(7471): 381-4, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24005329

ABSTRACT

Repair of interstrand crosslinks (ICLs) requires the coordinated action of the intra-S-phase checkpoint and the Fanconi anaemia pathway, which promote ICL incision, translesion synthesis and homologous recombination (reviewed in refs 1, 2). Previous studies have implicated the 3'-5' superfamily 2 helicase HELQ in ICL repair in Drosophila melanogaster (MUS301 (ref. 3)) and Caenorhabditis elegans (HELQ-1 (ref. 4)). Although in vitro analysis suggests that HELQ preferentially unwinds synthetic replication fork substrates with 3' single-stranded DNA overhangs and also disrupts protein-DNA interactions while translocating along DNA, little is known regarding its functions in mammalian organisms. Here we report that HELQ helicase-deficient mice exhibit subfertility, germ cell attrition, ICL sensitivity and tumour predisposition, with Helq heterozygous mice exhibiting a similar, albeit less severe, phenotype than the null, indicative of haploinsufficiency. We establish that HELQ interacts directly with the RAD51 paralogue complex BCDX2 and functions in parallel to the Fanconi anaemia pathway to promote efficient homologous recombination at damaged replication forks. Thus, our results reveal a critical role for HELQ in replication-coupled DNA repair, germ cell maintenance and tumour suppression in mammals.


Subject(s)
Carcinogenesis , DNA Helicases/metabolism , DNA Repair , Germ Cells/metabolism , Germ Cells/pathology , Rad51 Recombinase/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , DNA Damage/genetics , DNA Helicases/deficiency , DNA Helicases/genetics , DNA Repair/genetics , DNA Replication/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Gene Deletion , Germ Cells/cytology , Male , Mice , Multiprotein Complexes/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Recombinational DNA Repair/genetics
10.
Gastroenterology ; 151(3): 526-39, 2016 09.
Article in English | MEDLINE | ID: mdl-27215660

ABSTRACT

BACKGROUND & AIMS: Pancreatitis is the most important risk factor for pancreatic ductal adenocarcinoma (PDAC). Pancreatitis predisposes to PDAC because it induces a process of acinar cell reprogramming known as acinar-to-ductal metaplasia (ADM)-a precursor of pancreatic intraepithelial neoplasia lesions that can progress to PDAC. Mutations in KRAS are found at the earliest stages of pancreatic tumorigenesis, and it appears to be a gatekeeper to cancer progression. We investigated how mutations in KRAS cooperate with pancreatitis to promote pancreatic cancer progression in mice. METHODS: We generated mice carrying conditional alleles of Yap1 and Taz and disrupted Yap1 and Taz using a Cre-lox recombination strategy in adult mouse pancreatic acinar cells (Yap1fl/fl;Tazfl/fl;Ela1-CreERT2). We crossed these mice with LSL-KrasG12D mice, which express a constitutively active form of KRAS after Cre recombination. Pancreatic tumor initiation and progression were analyzed after chemically induced pancreatitis. We analyzed pancreatic tissues from patients with pancreatitis or PDAC by immunohistochemistry. RESULTS: Oncogenic activation of KRAS in normal, untransformed acinar cells in the pancreatic tissues of mice resulted in increased levels of pancreatitis-induced ADM. Expression of the constitutive active form of KRAS in this system led to activation of the transcriptional regulators YAP1 and TAZ; their function was required for pancreatitis-induced ADM in mice. The JAK-STAT3 pathway was a downstream effector of KRAS signaling via YAP1 and TAZ. YAP1 and TAZ directly mediated transcriptional activation of several genes in the JAK-STAT3 signaling pathway; this could be a mechanism by which acinar cells that express activated KRAS become susceptible to inflammation. CONCLUSIONS: We identified a mechanism by which oncogenic KRAS facilitates ADM and thereby generates the cells that initiate neoplastic progression. This process involves activation of YAP1 and TAZ in acinar cells, which up-regulate JAK-STAT3 signaling to promote development of PDAC in mice.


Subject(s)
Carcinogenesis/genetics , Pancreatic Neoplasms/genetics , Pancreatitis/complications , Signal Transduction/genetics , Acinar Cells/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Janus Kinases/metabolism , Mice , Mutation , Pancreas/pathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Pancreatitis/pathology , Phosphoproteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Risk Factors , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Up-Regulation , YAP-Signaling Proteins
11.
J Pathol ; 237(2): 152-65, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25965974

ABSTRACT

Metabolic reprogramming in cancer enhances macromolecule biosynthesis and supports cell survival. Oncogenic drivers affect metabolism by altering distinct metabolic processes and render cancer cells sensitive to perturbations of the metabolic network. This study aimed to identify selective metabolic dependencies in breast cancer by investigating 17 breast cancer cells lines representative of the genetic diversity of the disease. Using a functional screen, we demonstrate here that monocarboxylate transporter 4 (MCT4) is an important regulator of breast cancer cell survival. MCT4 supports pH maintenance, lactate secretion and non-oxidative glucose metabolism in breast cancer cells. Moreover, MCT4 depletion caused an increased dependence of cancer cells on mitochondrial respiration and glutamine metabolism. MCT4 depletion reduced the ability of breast cancer cells to grow in a three-dimensional (3D) matrix or as multilayered spheroids. Moreover, MCT4 expression is regulated by the PI3K-Akt signalling pathway and highly expressed in HER2-positive breast cancers. These results suggest that MCT4 is a potential therapeutic target in defined breast cancer subtypes and reveal novel avenues for combination treatment.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Energy Metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation , Cell Survival , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Humans , Hydrogen-Ion Concentration , Lactic Acid/metabolism , MCF-7 Cells , Mice, Nude , Monocarboxylic Acid Transporters/genetics , Muscle Proteins/genetics , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, ErbB-2/metabolism , Signal Transduction , Spheroids, Cellular , Time Factors , Transfection , Tumor Burden
12.
Ann Surg ; 261(3): 473-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25243543

ABSTRACT

OBJECTIVE: This study aimed to determine the prognostic significance of extramural venous invasion (EMVI) after chemoradiotherapy (CRT) by both magnetic resonance imaging (MRI) (ymrEMVI) and histopathology (ypEMVI). BACKGROUND: EMVI is a prognostic factor in rectal cancer but whether this remains so after CRT preoperative is unknown. Histopathological definitions of EMVI are variable and lead to underreporting particularly after CRT. METHODS: All consecutive patients staged on initial MRI as EMVI-positive undergoing preoperative CRT and curative surgery between Jan 2006 and Jan 2012 were included. Posttreatment EMVI status (yEMVI) was reevaluated for both MRI and pathology. The primary endpoint of disease-free survival (DFS) for ymrEMVI and ypEMVI was calculated using the Kaplan-Meier product limit and compared with a Mantel-Cox log-rank test. A P < 0.05 was considered significant. Hazard ratios (HRs) for disease recurrence were generated using Cox proportional hazard regression for MRI and histopathology tumor characteristics. RESULTS: A total of 188 patients who had evidence of EMVI on initial baseline MRI staging were included. MRI detected significantly more patients with persistent EMVI than histopathology (53% vs 19%) but both were prognostic for worse survival-ymrEMVI (HR 1.97) and ypEMVI (HR 2.39). Patients with persistent ymrEMVI-positivity had significantly worse DFS at 3 years (42.7%) compared with ymrEMVI-negative tumors (79.8%); DFS for was 36.9% versus 65.9% positive and negative ypEMVI, respectively. CONCLUSIONS: Detection of EMVI post-CRT is prognostically significant whether detected by MRI or histopathology. EMVI status after treatment may be used to counsel patients regarding ongoing risks of metastatic disease, implications for surveillance, and systemic chemotherapy.


Subject(s)
Chemoradiotherapy , Magnetic Resonance Imaging/methods , Neoplasm Invasiveness/pathology , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , Vascular Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Treatment Outcome
13.
N Engl J Med ; 366(10): 883-892, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22397650

ABSTRACT

BACKGROUND: Intratumor heterogeneity may foster tumor evolution and adaptation and hinder personalized-medicine strategies that depend on results from single tumor-biopsy samples. METHODS: To examine intratumor heterogeneity, we performed exome sequencing, chromosome aberration analysis, and ploidy profiling on multiple spatially separated samples obtained from primary renal carcinomas and associated metastatic sites. We characterized the consequences of intratumor heterogeneity using immunohistochemical analysis, mutation functional analysis, and profiling of messenger RNA expression. RESULTS: Phylogenetic reconstruction revealed branched evolutionary tumor growth, with 63 to 69% of all somatic mutations not detectable across every tumor region. Intratumor heterogeneity was observed for a mutation within an autoinhibitory domain of the mammalian target of rapamycin (mTOR) kinase, correlating with S6 and 4EBP phosphorylation in vivo and constitutive activation of mTOR kinase activity in vitro. Mutational intratumor heterogeneity was seen for multiple tumor-suppressor genes converging on loss of function; SETD2, PTEN, and KDM5C underwent multiple distinct and spatially separated inactivating mutations within a single tumor, suggesting convergent phenotypic evolution. Gene-expression signatures of good and poor prognosis were detected in different regions of the same tumor. Allelic composition and ploidy profiling analysis revealed extensive intratumor heterogeneity, with 26 of 30 tumor samples from four tumors harboring divergent allelic-imbalance profiles and with ploidy heterogeneity in two of four tumors. CONCLUSIONS: Intratumor heterogeneity can lead to underestimation of the tumor genomics landscape portrayed from single tumor-biopsy samples and may present major challenges to personalized-medicine and biomarker development. Intratumor heterogeneity, associated with heterogeneous protein function, may foster tumor adaptation and therapeutic failure through Darwinian selection. (Funded by the Medical Research Council and others.).


Subject(s)
Carcinoma, Renal Cell/genetics , Evolution, Molecular , Genetic Heterogeneity , Kidney Neoplasms/genetics , Phenotype , Biomarkers, Tumor , Biopsy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/secondary , Chromosome Aberrations , Everolimus , Exome , Genetic Heterogeneity/drug effects , Humans , Immunosuppressive Agents/pharmacology , Kidney/pathology , Kidney Neoplasms/pathology , Mutation , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Phylogeny , Ploidies , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
14.
Genome Res ; 22(2): 196-207, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22183965

ABSTRACT

Next generation sequencing has enabled systematic discovery of mutational spectra in cancer samples. Here, we used whole genome sequencing to characterize somatic mutations and structural variation in a primary acral melanoma and its lymph node metastasis. Our data show that the somatic mutational rates in this acral melanoma sample pair were more comparable to the rates reported in cancer genomes not associated with mutagenic exposure than in the genome of a melanoma cell line or the transcriptome of melanoma short-term cultures. Despite the perception that acral skin is sun-protected, the dominant mutational signature in these samples is compatible with damage due to ultraviolet light exposure. A nonsense mutation in ERCC5 discovered in both the primary and metastatic tumors could also have contributed to the mutational signature through accumulation of unrepaired dipyrimidine lesions. However, evidence of transcription-coupled repair was suggested by the lower mutational rate in the transcribed regions and expressed genes. The primary and the metastasis are highly similar at the level of global gene copy number alterations, loss of heterozygosity and single nucleotide variation (SNV). Furthermore, the majority of the SNVs in the primary tumor were propagated in the metastasis and one nonsynonymous coding SNV and one splice site mutation appeared to arise de novo in the metastatic lesion.


Subject(s)
Genome, Human , High-Throughput Nucleotide Sequencing , Melanoma/genetics , Aged , DNA Copy Number Variations , DNA Mutational Analysis , Exome , Humans , Loss of Heterozygosity , Male , Melanoma/pathology , Mutation Rate , Neoplasm Metastasis , Polymorphism, Single Nucleotide
15.
Cancer Cell ; 11(4): 311-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17418408

ABSTRACT

Germline mutations in the fumarate hydratase (FH) tumor suppressor gene predispose to leiomyomatosis, renal cysts, and renal cell cancer (HLRCC). HLRCC tumors overexpress HIF1alpha and hypoxia pathway genes. We conditionally inactivated mouse Fh1 in the kidney. Fh1 mutants developed multiple clonal renal cysts that overexpressed Hif1alpha and Hif2alpha. Hif targets, such as Glut1 and Vegf, were upregulated. We found that Fh1-deficient murine embryonic stem cells and renal carcinomas from HLRCC showed similar overexpression of HIF and hypoxia pathway components to the mouse cysts. Our data have shown in vivo that pseudohypoxic drive, resulting from HIF1alpha (and HIF2alpha) overexpression, is a direct consequence of Fh1 inactivation. Our mouse may be useful for testing therapeutic interventions that target angiogenesis and HIF-prolyl hydroxylation.


Subject(s)
Carcinoma, Renal Cell/etiology , Fumarate Hydratase/genetics , Gene Silencing/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Diseases, Cystic/etiology , Kidney Neoplasms/etiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Hypoxia , Cell Proliferation , Female , Glucose Transporter Type 1/metabolism , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
16.
J Pathol ; 230(3): 261-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23620124

ABSTRACT

Mucosal melanoma displays distinct clinical and epidemiological features compared to cutaneous melanoma. Here we used whole genome and whole exome sequencing to characterize the somatic alterations and mutation spectra in the genomes of ten mucosal melanomas. We observed somatic mutation rates that are considerably lower than occur in sun-exposed cutaneous melanoma, but comparable to the rates seen in cancers not associated with exposure to known mutagens. In particular, the mutation signatures are not indicative of ultraviolet light- or tobacco smoke-induced DNA damage. Genes previously reported as mutated in other cancers were also mutated in mucosal melanoma. Notably, there were substantially more copy number and structural variations in mucosal melanoma than have been reported in cutaneous melanoma. Thus, mucosal and cutaneous melanomas are distinct diseases with discrete genetic features. Our data suggest that different mechanisms underlie the genesis of these diseases and that structural variations play a more important role in mucosal than in cutaneous melanomagenesis.


Subject(s)
Esophageal Neoplasms/genetics , Genital Neoplasms, Female/genetics , Genital Neoplasms, Male/genetics , Melanoma/genetics , Paranasal Sinus Neoplasms/genetics , Rectal Neoplasms/genetics , Base Sequence , DNA Copy Number Variations , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Esophageal Neoplasms/pathology , Exome , Female , Genital Neoplasms, Female/pathology , Genital Neoplasms, Male/pathology , Genomics , High-Throughput Nucleotide Sequencing , Humans , Male , Melanoma/pathology , Molecular Sequence Data , Mucous Membrane/pathology , Mutation , Mutation Rate , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Paranasal Sinus Neoplasms/pathology , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins B-raf/genetics , Rectal Neoplasms/pathology , Sequence Analysis, DNA , Sequence Analysis, RNA , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/secondary
17.
J Pathol ; 231(3): 301-10, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24308032

ABSTRACT

The incidence of oesophagogastric junctional (OGJ) adenocarcinoma is rising rapidly in western countries, in contrast to the declining frequency of distal gastric carcinoma. Treatment options for adenocarcinomas involving the oesophagogastric junction are limited and the overall prognosis is extremely poor. To determine the genomic landscape of OGJ adenocarcinoma, exomes of eight tumours and matched germline DNA were subjected to massively parallel DNA sequencing. Microsatellite instability was observed in three tumours which coincided with an elevated number of somatic mutations. In total, 117 genes were identified that had predicted coding alterations in more than one tumour. Potentially actionable coding mutations were identified in 67 of these genes, including those in CR2, HGF , FGFR4, and ESRRB. Twenty-nine genes harbouring somatic coding mutations and copy number changes in the MSS OGJ dataset are also known to be altered with similar predicted functional consequence in other tumour types. Compared with the published mutational profile of gastric cancers, 49% (57/117) of recurrently mutated genes were unique to OGJ tumours. TP53, SYNE1, and ARID1A were amongst the most frequently mutated genes in a larger OGJ cohort. Our study provides an insight into the mutational landscape of OGJ adenocarcinomas and confirms that this is a highly mutated and heterogeneous disease. Furthermore, we have uncovered somatic mutations in therapeutically relevant genes which may represent candidate drug targets.


Subject(s)
Adenocarcinoma/genetics , DNA, Neoplasm/genetics , Esophageal Neoplasms/genetics , Esophagogastric Junction , Mutation , Stomach Neoplasms/genetics , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Adenosine Triphosphatases/analysis , Adenosine Triphosphatases/genetics , Adult , Aged , DNA Copy Number Variations/genetics , DNA Mutational Analysis , DNA Repair Enzymes/analysis , DNA Repair Enzymes/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Esophageal Neoplasms/chemistry , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Exome/genetics , Female , Genome, Human/genetics , Humans , Immunohistochemistry , Loss of Heterozygosity/genetics , Male , Microsatellite Instability , Middle Aged , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutL Proteins , Mutation/genetics , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Neoplasm Staging , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Polymerase Chain Reaction/methods , Prospective Studies , Stomach Neoplasms/chemistry , Stomach Neoplasms/pathology
18.
Cell Rep Med ; 5(3): 101435, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38417447

ABSTRACT

Mucosal (MM) and acral melanomas (AM) are rare melanoma subtypes of unmet clinical need; 15%-20% harbor KIT mutations potentially targeted by small-molecule inhibitors, but none yet approved in melanoma. This multicenter, single-arm Phase II trial (NICAM) investigates nilotinib safety and activity in KIT mutated metastatic MM and AM. KIT mutations are identified in 39/219 screened patients (18%); of 29/39 treated, 26 are evaluable for primary analysis. Six patients were alive and progression free at 6 months (local radiology review, 25%); 5/26 (19%) had objective response at 12 weeks; median OS was 7.7 months; ddPCR assay correctly identifies KIT alterations in circulating tumor DNA (ctDNA) in 16/17 patients. Nilotinib is active in KIT-mutant AM and MM, comparable to other KIT inhibitors, with demonstrable activity in nonhotspot KIT mutations, supporting broadening of KIT evaluation in AM and MM. Our results endorse further investigations of nilotinib for the treatment of KIT-mutated melanoma. This clinical trial was registered with ISRCTN (ISRCTN39058880) and EudraCT (2009-012945-49).


Subject(s)
Antineoplastic Agents , Melanoma , Skin Neoplasms , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Antineoplastic Agents/adverse effects , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Pyrimidines/adverse effects
19.
J Pathol ; 226(1): 73-83, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22009253

ABSTRACT

Adenomatous polyposis coli (APC ) mutations are found in most colorectal tumours. These mutations are almost always protein-truncating, deleting both central domains that regulate Wnt signalling and C-terminal domains that interact with the cytoskeleton. The importance of Wnt dysregulation for colorectal tumourigenesis is well characterized. It is, however, unclear whether loss of C-terminal functions contributes to tumourigenesis, although this protein region has been implicated in cellular processes--including polarity, migration, mitosis, and chromosomal instability (CIN)­that have been postulated as critical for the development and progression of intestinal tumours. Since almost all APC mutations in human patients disrupt both central and C-terminal regions, we created a mouse model to test the role of the C-terminus of APC in intestinal tumourigenesis. This mouse (Apc(ΔSAMP)) carries an internal deletion within Apc that dysregulates Wnt by removing the beta-catenin-binding and SAMP repeats, but leaves the C-terminus intact. We compared Apc(ΔSAMP) mice with Apc(1322T) animals. The latter allele represented the most commonly found human APC mutation and was identical to Apc(ΔSAMP) except for absence of the entire C-terminus. Apc(ΔSAMP) mice developed numerous intestinal adenomas indistinguishable in number, location, and dysplasia from those seen in Apc(1322T) mice. No carcinomas were found in Apc(ΔSAMP) or Apc(1322T) animals. While similar disruption of the Wnt signalling pathway was observed in tumours from both mice, no evidence of differential C-terminus functions (such as cell migration, CIN, or localization of APC and EB1) was seen. We conclude that the C-terminus of APC does not influence intestinal adenoma development or progression.


Subject(s)
Adenoma/genetics , Adenomatous Polyposis Coli Protein/genetics , Intestinal Neoplasms/genetics , Adenoma/pathology , Adenomatous Polyposis Coli Protein/chemistry , Animals , Blotting, Western , Cell Movement/genetics , Disease Progression , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization , Intestinal Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary/genetics , Signal Transduction/genetics , Wnt Signaling Pathway
20.
J Pathol ; 227(2): 146-56, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22362593

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common pathological subtype of kidney cancer. Here, we integrated an unbiased genome-wide RNA interference screen for ccRCC survival regulators with an analysis of recurrently overexpressed genes in ccRCC to identify new therapeutic targets in this disease. One of the most potent survival regulators, the monocarboxylate transporter MCT4 (SLC16A3), impaired ccRCC viability in all eight ccRCC lines tested and was the seventh most overexpressed gene in a meta-analysis of five ccRCC expression datasets. MCT4 silencing impaired secretion of lactate generated through glycolysis and induced cell cycle arrest and apoptosis. Silencing MCT4 resulted in intracellular acidosis, and reduction in intracellular ATP production together with partial reversion of the Warburg effect in ccRCC cell lines. Intra-tumoural heterogeneity in the intensity of MCT4 protein expression was observed in primary ccRCCs. MCT4 protein expression analysis based on the highest intensity of expression in primary ccRCCs was associated with poorer relapse-free survival, whereas modal intensity correlated with Fuhrman nuclear grade. Consistent with the potential selection of subclones enriched for MCT4 expression during disease progression, MCT4 expression was greater at sites of metastatic disease. These data suggest that MCT4 may serve as a novel metabolic target to reverse the Warburg effect and limit disease progression in ccRCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Glycolysis/genetics , Kidney Neoplasms/genetics , Monocarboxylic Acid Transporters/genetics , Muscle Proteins/genetics , RNA Interference , Apoptosis , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease-Free Survival , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Hydrogen-Ion Concentration , Kaplan-Meier Estimate , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Lactic Acid/metabolism , Phenotype , Prognosis , RNA, Messenger/metabolism , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL