Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nat Biotechnol ; 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862616

ABSTRACT

Subclonal reconstruction algorithms use bulk DNA sequencing data to quantify parameters of tumor evolution, allowing an assessment of how cancers initiate, progress and respond to selective pressures. We launched the ICGC-TCGA (International Cancer Genome Consortium-The Cancer Genome Atlas) DREAM Somatic Mutation Calling Tumor Heterogeneity and Evolution Challenge to benchmark existing subclonal reconstruction algorithms. This 7-year community effort used cloud computing to benchmark 31 subclonal reconstruction algorithms on 51 simulated tumors. Algorithms were scored on seven independent tasks, leading to 12,061 total runs. Algorithm choice influenced performance substantially more than tumor features but purity-adjusted read depth, copy-number state and read mappability were associated with the performance of most algorithms on most tasks. No single algorithm was a top performer for all seven tasks and existing ensemble strategies were unable to outperform the best individual methods, highlighting a key research need. All containerized methods, evaluation code and datasets are available to support further assessment of the determinants of subclonal reconstruction accuracy and development of improved methods to understand tumor evolution.

2.
Bioinformatics ; 40(5)2024 May 02.
Article in English | MEDLINE | ID: mdl-38662553

ABSTRACT

SUMMARY: Existing clustering methods for characterizing cell populations from single-cell RNA sequencing are constrained by several limitations stemming from the fact that clusters often cannot be homogeneous, particularly for transitioning populations. On the other hand, dominant cell populations within samples can be identified independently by their strong gene co-expression signatures using methods unrelated to partitioning. Here, we introduce a clustering method, CASCC (co-expression-assisted single-cell clustering), designed to improve biological accuracy using gene co-expression features identified using an unsupervised adaptive attractor algorithm. CASCC outperformed other methods as evidenced by multiple evaluation metrics, and our results suggest that CASCC can improve the analysis of single-cell transcriptomics, enabling potential new discoveries related to underlying biological mechanisms. AVAILABILITY AND IMPLEMENTATION: The CASCC R package is publicly available at https://github.com/LingyiC/CASCC and https://zenodo.org/doi/10.5281/zenodo.10648327.


Subject(s)
Algorithms , RNA-Seq , Single-Cell Analysis , Software , Single-Cell Analysis/methods , Cluster Analysis , RNA-Seq/methods , Humans , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Gene Expression Analysis
3.
Cancer Res ; 84(5): 648-649, 2024 03 04.
Article in English | MEDLINE | ID: mdl-38437636

ABSTRACT

Cancer aggressiveness has been linked with obesity, and studies have shown that adipose tissue can enhance cancer progression. In this issue of Cancer Research, Hosni and colleagues discover a paracrine mechanism mediated by adipocyte precursor cells through which urothelial carcinomas become resistant to erdafitinib, a recently approved therapy inhibiting fibroblast growth factor receptors (FGFR). They identified neuregulin 1 (NRG1) secreted by adipocyte precursor cells as an activator of HER3 signaling that enables resistance. The NRG1-mediated FGFR inhibitor resistance was amenable to intervention with pertuzumab, an antibody blocking the NRG1/HER3 axis. To investigate the nature of the resistance-associated NRG1-expressing cells in human patients, the authors analyzed published single-cell RNA sequencing data and observed that such cells appear in a cluster assigned as inflammatory cancer-associated fibroblasts (iCAF). Notably, the gene signature corresponding to these CAFs is highly similar to that shared by adipose stromal cells (ASC) in fat tissue and fibro-adipogenic progenitors (FAP) in skeletal muscle of cancer-free individuals. Because fibroblasts with the ASC/FAP signature are enriched in various carcinomas, it is possible that the paracrine signaling conferred by NRG1 is a pan-cancer mechanism of FGFR inhibitor resistance and tumor aggressiveness. See related article by Hosni et al., p. 725.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Transitional Cell , Humans , Adipocytes , Adipose Tissue , Stromal Cells
4.
Article in English | MEDLINE | ID: mdl-38466528

ABSTRACT

We identified a progenitor cell population highly enriched in samples from invasive and chemo-resistant carcinomas, characterized by a well-defined multigene signature including APOD, DCN, and LUM. This cell population has previously been labeled as consisting of inflammatory cancer-associated fibroblasts (iCAFs). The same signature characterizes naturally occurring fibro-adipogenic progenitors (FAPs) as well as stromal cells abundant in normal adipose tissue. Our analysis of human gene expression databases provides evidence that adipose stromal cells (ASCs) are recruited by tumors and undergo differentiation into CAFs during cancer progression to invasive and chemotherapy-resistant stages.

5.
PLoS Comput Biol ; 17(7): e1009228, 2021 07.
Article in English | MEDLINE | ID: mdl-34283835

ABSTRACT

During the last ten years, many research results have been referring to a particular type of cancer-associated fibroblasts associated with poor prognosis, invasiveness, metastasis and resistance to therapy in multiple cancer types, characterized by a gene expression signature with prominent presence of genes COL11A1, THBS2 and INHBA. Identifying the underlying biological mechanisms responsible for their creation may facilitate the discovery of targets for potential pan-cancer therapeutics. Using a novel computational approach for single-cell gene expression data analysis identifying the dominant cell populations in a sequence of samples from patients at various stages, we conclude that these fibroblasts are produced by a pan-cancer cellular transition originating from a particular type of adipose-derived stromal cells naturally present in the stromal vascular fraction of normal adipose tissue, having a characteristic gene expression signature. Focusing on a rich pancreatic cancer dataset, we provide a detailed description of the continuous modification of the gene expression profiles of cells as they transition from APOD-expressing adipose-derived stromal cells to COL11A1-expressing cancer-associated fibroblasts, identifying the key genes that participate in this transition. These results also provide an explanation to the well-known fact that the adipose microenvironment contributes to cancer progression.


Subject(s)
Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts/metabolism , Collagen Type XI/genetics , Neoplasm Invasiveness/genetics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Computational Biology , Databases, Factual , Databases, Genetic , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Single-Cell Analysis , Stromal Cells/metabolism , Stromal Cells/pathology , Transcriptome , Tumor Microenvironment/genetics
6.
IEEE/ACM Trans Comput Biol Bioinform ; 18(6): 2271-2280, 2021.
Article in English | MEDLINE | ID: mdl-32070995

ABSTRACT

Bulk samples of the same patient are heterogeneous in nature, comprising of different subpopulations (subclones) of cancer cells. Cells in a tumor subclone are characterized by unique mutational genotype profile. Resolving tumor heterogeneity by estimating the genotypes, cellular proportions and the number of subclones present in the tumor can help in understanding cancer progression and treatment. We present a novel method, ChaClone2, to efficiently deconvolve the observed variant allele fractions (VAFs), with consideration for possible effects from copy number aberrations at the mutation loci. Our method describes a state-space formulation of the feature allocation model, deconvolving the observed VAFs from samples of the same patient into three matrices: subclonal total and variant copy numbers for mutated genes, and proportions of subclones in each sample. We describe an efficient sequential Monte Carlo (SMC) algorithm to estimate these matrices. Extensive simulation shows that the ChaClone2 yields better accuracy when compared with other state-of-the-art methods for addressing similar problem and it offers scalability to large datasets. Also, ChaClone2 features that the model parameter estimates can be refined whenever new mutation data of freshly sequenced genomic locations are available. MATLAB code and datasets are available to download at: https://github.com/moyanre/method2.


Subject(s)
Computational Biology/methods , DNA Copy Number Variations/genetics , Mutation/genetics , Neoplasms/genetics , Algorithms , Bayes Theorem , Genetic Heterogeneity , Humans , Monte Carlo Method , Stochastic Processes
7.
Sci Rep ; 10(1): 17199, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057153

ABSTRACT

Analysis of large gene expression datasets from biopsies of cancer patients can identify co-expression signatures representing particular biomolecular events in cancer. Some of these signatures involve genomically co-localized genes resulting from the presence of copy number alterations (CNAs), for which analysis of the expression of the underlying genes provides valuable information about their combined role as oncogenes or tumor suppressor genes. Here we focus on the discovery and interpretation of such signatures that are present in multiple cancer types due to driver amplifications and deletions in particular regions of the genome after doing a comprehensive analysis combining both gene expression and CNA data from The Cancer Genome Atlas.


Subject(s)
DNA Copy Number Variations/genetics , Neoplasms/genetics , Oncogenes/genetics , Data Analysis , Gene Dosage/genetics , Gene Expression/genetics , Genomics/methods , Humans
8.
Bioinformatics ; 36(11): 3588-3589, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32108864

ABSTRACT

SUMMARY: We developed 2DImpute, an imputation method for correcting false zeros (known as dropouts) in single-cell RNA-sequencing (scRNA-seq) data. It features preventing excessive correction by predicting the false zeros and imputing their values by making use of the interrelationships between both genes and cells in the expression matrix. We showed that 2DImpute outperforms several leading imputation methods by applying it on datasets from various scRNA-seq protocols. AVAILABILITY AND IMPLEMENTATION: The R package of 2DImpute is freely available at GitHub (https://github.com/zky0708/2DImpute). CONTACT: d.anastassiou@columbia.edu. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
RNA-Seq , Software , Sequence Analysis, RNA , Single-Cell Analysis , Exome Sequencing
9.
Nat Biotechnol ; 38(1): 97-107, 2020 01.
Article in English | MEDLINE | ID: mdl-31919445

ABSTRACT

Tumor DNA sequencing data can be interpreted by computational methods that analyze genomic heterogeneity to infer evolutionary dynamics. A growing number of studies have used these approaches to link cancer evolution with clinical progression and response to therapy. Although the inference of tumor phylogenies is rapidly becoming standard practice in cancer genome analyses, standards for evaluating them are lacking. To address this need, we systematically assess methods for reconstructing tumor subclonality. First, we elucidate the main algorithmic problems in subclonal reconstruction and develop quantitative metrics for evaluating them. Then we simulate realistic tumor genomes that harbor all known clonal and subclonal mutation types and processes. Finally, we benchmark 580 tumor reconstructions, varying tumor read depth, tumor type and somatic variant detection. Our analysis provides a baseline for the establishment of gold-standard methods to analyze tumor heterogeneity.


Subject(s)
Algorithms , Neoplasms/pathology , Clone Cells , Computer Simulation , DNA Copy Number Variations/genetics , Gene Dosage , Genome , Humans , Mutation/genetics , Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Reference Standards
11.
PLoS One ; 14(1): e0211213, 2019.
Article in English | MEDLINE | ID: mdl-30682127

ABSTRACT

Tumors are heterogeneous in the sense that they consist of multiple subpopulations of cells, referred to as subclones, each of which is characterized by a distinct profile of genomic variations such as somatic mutations. Inferring the underlying clonal landscape has become an important topic in that it can help in understanding cancer development and progression, and thereby help in improving treatment. We describe a novel state-space model, based on the feature allocation framework and an efficient sequential Monte Carlo (SMC) algorithm, using the somatic mutation data obtained from tumor samples to estimate the number of subclones, as well as their characterization. Our approach, by design, is capable of handling any number of mutations. Via extensive simulations, our method exhibits high accuracy, in most cases, and compares favorably with existing methods. Moreover, we demonstrated the validity of our method through analyzing real tumor samples from patients from multiple cancer types (breast, prostate, and lung). Our results reveal driver mutation events specific to cancer types, and indicate clonal expansion by manual phylogenetic analysis. MATLAB code and datasets are available to download at: https://github.com/moyanre/tumor_clones.


Subject(s)
Clone Cells/cytology , Mutation , Neoplasms/genetics , Algorithms , Cell Count , Clone Cells/chemistry , Disease Progression , Genotype , Humans , Models, Theoretical , Monte Carlo Method , Phylogeny
12.
Transl Psychiatry ; 9(1): 32, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670680

ABSTRACT

Similar environmental risk factors have been implicated in different neuropsychiatric disorders (including major psychiatric and neurodegenerative diseases), indicating the existence of common epigenetic mechanisms underlying the pathogenesis shared by different illnesses. To investigate such commonality, we applied an unsupervised computational approach identifying several consensus co-expression and co-methylation signatures from a data cohort of postmortem prefrontal cortex (PFC) samples from individuals with six different neuropsychiatric disorders-schizophrenia, bipolar disorder, major depression, alcoholism, Alzheimer's and Parkinson's-as well as healthy controls. Among our results, we identified a pair of strongly interrelated co-expression and co-methylation (E-M) signatures showing consistent and significant disease association in multiple types of disorders. This E-M signature was enriched for interneuron markers, and we further demonstrated that it is unlikely for this enrichment to be due to varying subpopulation abundance of normal interneurons across samples. Moreover, gene set enrichment analysis revealed overrepresentation of stress-related biological processes in this E-M signature. Our integrative analysis of expression and methylation profiles, therefore, suggests a stress-related epigenetic mechanism in the brain, which could be associated with the pathogenesis of multiple neuropsychiatric diseases.


Subject(s)
Alcoholism/genetics , Alzheimer Disease/genetics , Bipolar Disorder/genetics , DNA Methylation , Depressive Disorder, Major/genetics , Parkinson Disease/genetics , Schizophrenia/genetics , Epigenesis, Genetic , Gene Regulatory Networks , Humans
13.
Immunity ; 48(4): 812-830.e14, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29628290

ABSTRACT

We performed an extensive immunogenomic analysis of more than 10,000 tumors comprising 33 diverse cancer types by utilizing data compiled by TCGA. Across cancer types, we identified six immune subtypes-wound healing, IFN-γ dominant, inflammatory, lymphocyte depleted, immunologically quiet, and TGF-ß dominant-characterized by differences in macrophage or lymphocyte signatures, Th1:Th2 cell ratio, extent of intratumoral heterogeneity, aneuploidy, extent of neoantigen load, overall cell proliferation, expression of immunomodulatory genes, and prognosis. Specific driver mutations correlated with lower (CTNNB1, NRAS, or IDH1) or higher (BRAF, TP53, or CASP8) leukocyte levels across all cancers. Multiple control modalities of the intracellular and extracellular networks (transcription, microRNAs, copy number, and epigenetic processes) were involved in tumor-immune cell interactions, both across and within immune subtypes. Our immunogenomics pipeline to characterize these heterogeneous tumors and the resulting data are intended to serve as a resource for future targeted studies to further advance the field.


Subject(s)
Genomics/methods , Neoplasms , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Macrophages/immunology , Male , Middle Aged , Neoplasms/classification , Neoplasms/genetics , Neoplasms/immunology , Prognosis , Th1-Th2 Balance/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Wound Healing/genetics , Wound Healing/immunology , Young Adult
15.
PLoS One ; 11(12): e0167994, 2016.
Article in English | MEDLINE | ID: mdl-27992465

ABSTRACT

Exploring linkage disequilibrium (LD) patterns among the single nucleotide polymorphism (SNP) sites can improve the accuracy and cost-effectiveness of genomic association studies, whereby representative (tag) SNPs are identified to sufficiently represent the genomic diversity in populations. There has been considerable amount of effort in developing efficient algorithms to select tag SNPs from the growing large-scale data sets. Methods using the classical pairwise-LD and multi-locus LD measures have been proposed that aim to reduce the computational complexity and to increase the accuracy, respectively. The present work solves the tag SNP selection problem by efficiently balancing the computational complexity and accuracy, and improves the coverage in genomic diversity in a cost-effective manner. The employed algorithm makes use of mutual information to explore the multi-locus association between SNPs and can handle different data types and conditions. Experiments with benchmark HapMap data sets show comparable or better performance against the state-of-the-art algorithms. In particular, as a novel application, the genome-wide SNP tagging is performed in the 1000 Genomes Project data sets, and produced a well-annotated database of tagging variants that capture the common genotype diversity in 2,504 samples from 26 human populations. Compared to conventional methods, the algorithm requires as input only the genotype (or haplotype) sequences, can scale up to genome-wide analyses, and produces accurate solutions with more information-rich output, providing an improved platform for researchers towards the subsequent association studies.


Subject(s)
Algorithms , Chromosome Mapping/methods , Genome-Wide Association Study/methods , Polymorphism, Single Nucleotide , Base Sequence , Cluster Analysis , Databases, Genetic , Epistasis, Genetic , Expressed Sequence Tags , Genetic Association Studies , Haplotypes , Humans , Linkage Disequilibrium , Sequence Homology, Nucleic Acid
16.
Cancer Epidemiol Biomarkers Prev ; 23(12): 2850-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25249324

ABSTRACT

BACKGROUND: The winning model of the Sage Bionetworks/DREAM Breast Cancer Prognosis Challenge made use of several molecular features, called attractor metagenes, as well as another metagene defined by the average expression level of the two genes FGD3 and SUSD3. This is a follow-up study toward developing a breast cancer prognostic test derived from and improving upon that model. METHODS: We designed a feature selector facility calculating the prognostic scores of combinations of features, including those that we had used earlier, as well as those used in existing breast cancer biomarker assays, identifying the optimal selection of features for the test. RESULTS: The resulting test, called BCAM (Breast Cancer Attractor Metagenes), is universally applicable to all clinical subtypes and stages of breast cancer and does not make any use of breast cancer molecular subtype or hormonal status information, none of which provided additional prognostic value. BCAM is composed of several molecular features: the breast cancer-specific FGD3-SUSD3 metagene, four attractor metagenes present in multiple cancer types (CIN, MES, LYM, and END), three additional individual genes (CD68, DNAJB9, and CXCL12), tumor size, and the number of positive lymph nodes. CONCLUSIONS: Our analysis leads to the unexpected and remarkable suggestion that ER, PR, and HER2 status, or molecular subtype classification, do not provide additional prognostic value when the values of the FGD3-SUSD3 and attractor metagenes are taken into consideration. IMPACT: Our results suggest that BCAM's prognostic predictions show potential to outperform those resulting from existing breast cancer biomarker assays.


Subject(s)
Breast Neoplasms/genetics , Biomarkers, Tumor , Breast Neoplasms/mortality , Female , Gene Expression Profiling , Humans , Metagenomics , Prognosis , Survival Rate
17.
Article in English | MEDLINE | ID: mdl-24868199

ABSTRACT

Copy number variations (CNVs) are abundant in the human genome. They have been associated with complex traits in genome-wide association studies (GWAS) and expected to continue playing an important role in identifying the etiology of disease phenotypes. As a result of current high throughput whole-genome single-nucleotide polymorphism (SNP) arrays, we currently have datasets that simultaneously have integer copy numbers in CNV regions as well as SNP genotypes. At the same time, haplotypes that have been shown to offer advantages over genotypes in identifying disease traits even though available for SNP genotypes are largely not available for CNV/SNP data due to insufficient computational tools. We introduce a new framework for inferring haplotypes in CNV/SNP data using a sequential Monte Carlo sampling scheme 'Tree-Based Deterministic Sampling CNV' (TDSCNV). We compare our method with polyHap(v2.0), the only currently available software able to perform inference in CNV/SNP genotypes, on datasets of varying number of markers. We have found that both algorithms show similar accuracy but TDSCNV is an order of magnitude faster while scaling linearly with the number of markers and number of individuals and thus could be the method of choice for haplotype inference in such datasets. Our method is implemented in the TDSCNV package which is available for download at http://www.ee.columbia.edu/~anastas/tdscnv.

18.
Cancer Immunol Res ; 2(4): 301-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24764577

ABSTRACT

Janus kinase-2 (JAK2) supports breast cancer growth, and clinical trials testing JAK2 inhibitors are under way. In addition to the tumor epithelium, JAK2 is also expressed in other tissues including immune cells; whether the JAK2 mRNA levels in breast tumors correlate with outcomes has not been evaluated. Using a case-control design, JAK2 mRNA was measured in 223 archived breast tumors and associations with distant recurrence were evaluated by logistic regression. The frequency of correct pairwise comparisons of patient rankings based on JAK2 levels versus survival outcomes, the concordance index (CI), was evaluated using data from 2,460 patients in three cohorts. In the case-control study, increased JAK2 was associated with a decreasing risk of recurrence (multivariate P = 0.003, n = 223). Similarly, JAK2 was associated with a protective CI (<0.5) in the public cohorts: NETHERLANDS CI = 0.376, n = 295; METABRIC CI = 0.462, n = 1,981; OSLOVAL CI = 0.452, n = 184. Furthermore, JAK2 was strongly correlated with the favorable prognosis LYM metagene signature for infiltrating T cells (r = 0.5; P < 2 × 10(-16); n = 1,981) and with severe lymphocyte infiltration (P = 0.00003, n = 156). Moreover, the JAK1/2 inhibitor ruxolitinib potently inhibited the anti-CD3-dependent production of IFN-γ, a marker of the differentiation of Th cells along the tumor-inhibitory Th1 pathway. The potential for JAK2 inhibitors to interfere with the antitumor capacities of T cells should be evaluated.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/immunology , Gene Expression , Janus Kinase 2/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Case-Control Studies , Female , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , RNA, Messenger/genetics , Recurrence , Treatment Outcome
19.
BMC Bioinformatics ; 14: 270, 2013 Sep 08.
Article in English | MEDLINE | ID: mdl-24010487

ABSTRACT

BACKGROUND: DNA pooling constitutes a cost effective alternative in genome wide association studies. In DNA pooling, equimolar amounts of DNA from different individuals are mixed into one sample and the frequency of each allele in each position is observed in a single genotype experiment. The identification of haplotype frequencies from pooled data in addition to single locus analysis is of separate interest within these studies as haplotypes could increase statistical power and provide additional insight. RESULTS: We developed a method for maximum-parsimony haplotype frequency estimation from pooled DNA data based on the sparse representation of the DNA pools in a dictionary of haplotypes. Extensions to scenarios where data is noisy or even missing are also presented. The resulting method is first applied to simulated data based on the haplotypes and their associated frequencies of the AGT gene. We further evaluate our methodology on datasets consisting of SNPs from the first 7Mb of the HapMap CEU population. Noise and missing data were further introduced in the datasets in order to test the extensions of the proposed method. Both HIPPO and HAPLOPOOL were also applied to these datasets to compare performances. CONCLUSIONS: We evaluate our methodology on scenarios where pooling is more efficient relative to individual genotyping; that is, in datasets that contain pools with a small number of individuals. We show that in such scenarios our methodology outperforms state-of-the-art methods such as HIPPO and HAPLOPOOL.


Subject(s)
DNA/chemistry , Gene Frequency/genetics , Genomics/methods , Haplotypes/genetics , Algorithms , DNA/genetics , Databases, Genetic , HapMap Project , Humans , Polymorphism, Single Nucleotide/genetics
20.
Vasc Cell ; 5(1): 17, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-24066611

ABSTRACT

BACKGROUND: Anti-angiogenesis is a validated strategy to treat cancer, with efficacy in controlling both primary tumor growth and metastasis. The role of the Notch family of proteins in tumor angiogenesis is still emerging, but recent data suggest that Notch signaling may function in the physiologic response to loss of VEGF signaling, and thus participate in tumor adaptation to VEGF inhibitors. METHODS: We asked whether combining Notch and VEGF blockade would enhance suppression of tumor angiogenesis and growth, using the NGP neuroblastoma model. NGP tumors were engineered to express a Notch1 decoy construct, which restricts Notch signaling, and then treated with either the anti-VEGF antibody bevacizumab or vehicle. RESULTS: Combining Notch and VEGF blockade led to blood vessel regression, increasing endothelial cell apoptosis and disrupting pericyte coverage of endothelial cells. Combined Notch and VEGF blockade did not affect tumor weight, but did additively reduce tumor viability. CONCLUSIONS: Our results indicate that Notch and VEGF pathways play distinct but complementary roles in tumor angiogenesis, and show that concurrent blockade disrupts primary tumor vasculature and viability further than inhibition of either pathway alone.

SELECTION OF CITATIONS
SEARCH DETAIL
...