Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Bioinformatics ; 38(20): 4713-4719, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36000873

ABSTRACT

MOTIVATION: Tumours evolve as heterogeneous populations of cells, which may be distinguished by different genomic aberrations. The resulting intra-tumour heterogeneity plays an important role in cancer patient relapse and treatment failure, so that obtaining a clear understanding of each patient's tumour composition and evolutionary history is key for personalized therapies. Single-cell sequencing (SCS) now provides the possibility to resolve tumour heterogeneity at the highest resolution of individual tumour cells, but brings with it challenges related to the particular noise profiles of the sequencing protocols as well as the complexity of the underlying evolutionary process. RESULTS: By modelling the noise processes and allowing mutations to be lost or to reoccur during tumour evolution, we present a method to jointly call mutations in each cell, reconstruct the phylogenetic relationship between cells, and determine the locations of mutational losses and recurrences. Our Bayesian approach allows us to accurately call mutations as well as to quantify our certainty in such predictions. We show the advantages of allowing mutational loss or recurrence with simulated data and present its application to tumour SCS data. AVAILABILITY AND IMPLEMENTATION: SCIΦN is available at https://github.com/cbg-ethz/SCIPhIN. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genomics , Neoplasms , Bayes Theorem , Humans , Mutation , Neoplasms/genetics , Phylogeny , Software
2.
BMC Genomics ; 22(1): 592, 2021 Aug 04.
Article in English | MEDLINE | ID: mdl-34348664

ABSTRACT

BACKGROUND: Genetic aberrations in hepatocellular carcinoma (HCC) are well known, but the functional consequences of such aberrations remain poorly understood. RESULTS: Here, we explored the effect of defined genetic changes on the transcriptome, proteome and phosphoproteome in twelve tumors from an mTOR-driven hepatocellular carcinoma mouse model. Using Network-based Integration of multi-omiCS data (NetICS), we detected 74 'mediators' that relay via molecular interactions the effects of genetic and miRNA expression changes. The detected mediators account for the effects of oncogenic mTOR signaling on the transcriptome, proteome and phosphoproteome. We confirmed the dysregulation of the mediators YAP1, GRB2, SIRT1, HDAC4 and LIS1 in human HCC. CONCLUSIONS: This study suggests that targeting pathways such as YAP1 or GRB2 signaling and pathways regulating global histone acetylation could be beneficial in treating HCC with hyperactive mTOR signaling.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Pharmaceutical Preparations , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Transcriptome
3.
Cancers (Basel) ; 13(9)2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946379

ABSTRACT

Intra-tumour heterogeneity is the molecular hallmark of renal cancer, and the molecular tumour composition determines the treatment outcome of renal cancer patients. In renal cancer tumourigenesis, in general, different tumour clones evolve over time. We analysed intra-tumour heterogeneity and subclonal mutation patterns in 178 tumour samples obtained from 89 clear cell renal cell carcinoma patients. In an initial discovery phase, whole-exome and transcriptome sequencing data from paired tumour biopsies from 16 ccRCC patients were used to design a gene panel for follow-up analysis. In this second phase, 826 selected genes were targeted at deep coverage in an extended cohort of 89 patients for a detailed analysis of tumour heterogeneity. On average, we found 22 mutations per patient. Pairwise comparison of the two biopsies from the same tumour revealed that on average, 62% of the mutations in a patient were detected in one of the two samples. In addition to commonly mutated genes (VHL, PBRM1, SETD2 and BAP1), frequent subclonal mutations with low variant allele frequency (<10%) were observed in TP53 and in mucin coding genes MUC6, MUC16, and MUC3A. Of the 89 ccRCC tumours, 87 (~98%) harboured private mutations, occurring in only one of the paired tumour samples. Clonally exclusive pathway pairs were identified using the WES data set from 16 ccRCC patients. Our findings imply that shared and private mutations significantly contribute to the complexity of differential gene expression and pathway interaction and might explain the clonal evolution of different molecular renal cancer subgroups. Multi-regional sequencing is central for the identification of subclones within ccRCC.

4.
Blood ; 135(18): 1548-1559, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32181816

ABSTRACT

Clonal hematopoiesis (CH) is associated with age and an increased risk of myeloid malignancies, cardiovascular risk, and all-cause mortality. We tested for CH in a setting where hematopoietic stem cells (HSCs) of the same individual are exposed to different degrees of proliferative stress and environments, ie, in long-term survivors of allogeneic hematopoietic stem cell transplantation (allo-HSCT) and their respective related donors (n = 42 donor-recipient pairs). With a median follow-up time since allo-HSCT of 16 years (range, 10-32 years), we found a total of 35 mutations in 23 out of 84 (27.4%) study participants. Ten out of 42 donors (23.8%) and 13 out of 42 recipients (31%) had CH. CH was associated with older donor and recipient age. We identified 5 cases of donor-engrafted CH, with 1 case progressing into myelodysplastic syndrome in both donor and recipient. Four out of 5 cases showed increased clone size in recipients compared with donors. We further characterized the hematopoietic system in individuals with CH as follows: (1) CH was consistently present in myeloid cells but varied in penetrance in B and T cells; (2) colony-forming units (CFUs) revealed clonal evolution or multiple independent clones in individuals with multiple CH mutations; and (3) telomere shortening determined in granulocytes suggested ∼20 years of added proliferative history of HSCs in recipients compared with their donors, with telomere length in CH vs non-CH CFUs showing varying patterns. This study provides insight into the long-term behavior of the same human HSCs and respective CH development under different proliferative conditions.


Subject(s)
Clonal Hematopoiesis , Hematopoietic Stem Cell Transplantation/mortality , Hematopoietic Stem Cells/metabolism , Tissue Donors , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Clonal Evolution/genetics , Colony-Forming Units Assay , DNA Mutational Analysis , Female , Hematopoietic Stem Cells/cytology , Humans , Male , Middle Aged , Mutation , Prognosis , Telomere , Transplant Recipients , Transplantation, Homologous , Treatment Outcome , Young Adult
5.
Nature ; 566(7745): 553-557, 2019 02.
Article in English | MEDLINE | ID: mdl-30728496

ABSTRACT

A better understanding of the features that define the interaction between cancer cells and immune cells is important for the development of new cancer therapies1. However, focus is often given to interactions that occur within the primary tumour and its microenvironment, whereas the role of immune cells during cancer dissemination in patients remains largely uncharacterized2,3. Circulating tumour cells (CTCs) are precursors of metastasis in several types of cancer4-6, and are occasionally found within the bloodstream in association with non-malignant cells such as white blood cells (WBCs)7,8. The identity and function of these CTC-associated WBCs, as well as the molecular features that define the interaction between WBCs and CTCs, are unknown. Here we isolate and characterize individual CTC-associated WBCs, as well as corresponding cancer cells within each CTC-WBC cluster, from patients with breast cancer and from mouse models. We use single-cell RNA sequencing to show that in the majority of these cases, CTCs were associated with neutrophils. When comparing the transcriptome profiles of CTCs associated with neutrophils against those of CTCs alone, we detect a number of differentially expressed genes that outline cell cycle progression, leading to more efficient metastasis formation. Further, we identify cell-cell junction and cytokine-receptor pairs that define CTC-neutrophil clusters, representing key vulnerabilities of the metastatic process. Thus, the association between neutrophils and CTCs drives cell cycle progression within the bloodstream and expands the metastatic potential of CTCs, providing a rationale for targeting this interaction in treatment of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cell Cycle , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Neutrophils/pathology , Animals , Breast Neoplasms/therapy , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Exons/genetics , Female , Gene Expression Profiling , Humans , Intercellular Junctions , Mice , Mutation/genetics , Neoplasm Metastasis/genetics , Neoplastic Cells, Circulating/metabolism , Neutrophils/metabolism , Sequence Analysis, RNA , Exome Sequencing
6.
Brief Bioinform ; 20(3): 778-788, 2019 05 21.
Article in English | MEDLINE | ID: mdl-29272324

ABSTRACT

Molecular profiling of tumor biopsies plays an increasingly important role not only in cancer research, but also in the clinical management of cancer patients. Multi-omics approaches hold the promise of improving diagnostics, prognostics and personalized treatment. To deliver on this promise of precision oncology, appropriate bioinformatics methods for managing, integrating and analyzing large and complex data are necessary. Here, we discuss the specific requirements of bioinformatics methods and software that arise in the setting of clinical oncology, owing to a stricter regulatory environment and the need for rapid, highly reproducible and robust procedures. We describe the workflow of a molecular tumor board and the specific bioinformatics support that it requires, from the primary analysis of raw molecular profiling data to the automatic generation of a clinical report and its delivery to decision-making clinical oncologists. Such workflows have to various degrees been implemented in many clinical trials, as well as in molecular tumor boards at specialized cancer centers and university hospitals worldwide. We review these and more recent efforts to include other high-dimensional multi-omics patient profiles into the tumor board, as well as the state of clinical decision support software to translate molecular findings into treatment recommendations.


Subject(s)
Computational Biology , Medical Oncology , Precision Medicine , High-Throughput Nucleotide Sequencing , Humans
7.
Nat Commun ; 9(1): 5144, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30514897

ABSTRACT

Reconstructing the evolution of tumors is a key aspect towards the identification of appropriate cancer therapies. The task is challenging because tumors evolve as heterogeneous cell populations. Single-cell sequencing holds the promise of resolving the heterogeneity of tumors; however, it has its own challenges including elevated error rates, allelic drop-out, and uneven coverage. Here, we develop a new approach to mutation detection in individual tumor cells by leveraging the evolutionary relationship among cells. Our method, called SCIΦ, jointly calls mutations in individual cells and estimates the tumor phylogeny among these cells. Employing a Markov Chain Monte Carlo scheme enables us to reliably call mutations in each single cell even in experiments with high drop-out rates and missing data. We show that SCIΦ outperforms existing methods on simulated data and applied it to different real-world datasets, namely a whole exome breast cancer as well as a panel acute lymphoblastic leukemia dataset.


Subject(s)
Mutation/genetics , Neoplasms/genetics , Phylogeny , Single-Cell Analysis/methods , Algorithms , Alleles , Cell Lineage/genetics , Computational Biology , DNA, Neoplasm/genetics , Exome/genetics , Gene Frequency , Genetic Heterogeneity , Humans , Loss of Heterozygosity/genetics , Markov Chains , Monte Carlo Method , Sensitivity and Specificity
8.
BMC Med Inform Decis Mak ; 18(1): 89, 2018 10 29.
Article in English | MEDLINE | ID: mdl-30373609

ABSTRACT

BACKGROUND: Molecular precision oncology is an emerging practice to improve cancer therapy by decreasing the risk of choosing treatments that lack efficacy or cause adverse events. However, the challenges of integrating molecular profiling into routine clinical care are manifold. From a computational perspective these include the importance of a short analysis turnaround time, the interpretation of complex drug-gene and gene-gene interactions, and the necessity of standardized high-quality workflows. In addition, difficulties faced when integrating molecular diagnostics into clinical practice are ethical concerns, legal requirements, and limited availability of treatment options beyond standard of care as well as the overall lack of awareness of their existence. METHODS: To the best of our knowledge, we are the first group in Switzerland that established a workflow for personalized diagnostics based on comprehensive high-throughput sequencing of tumors at the clinic. Our workflow, named SwissMTB (Swiss Molecular Tumor Board), links genetic tumor alterations and gene expression to therapeutic options and clinical trial opportunities. The resulting treatment recommendations are summarized in a clinical report and discussed in a molecular tumor board at the clinic to support therapy decisions. RESULTS: Here we present results from an observational pilot study including 22 late-stage cancer patients. In this study we were able to identify actionable variants and corresponding therapies for 19 patients. Half of the patients were analyzed retrospectively. In two patients we identified resistance-associated variants explaining lack of therapy response. For five out of eleven patients analyzed before treatment the SwissMTB diagnostic influenced treatment decision. CONCLUSIONS: SwissMTB enables the analysis and clinical interpretation of large numbers of potentially actionable molecular targets. Thus, our workflow paves the way towards a more frequent use of comprehensive molecular diagnostics in Swiss hospitals.


Subject(s)
Neoplasms/diagnosis , Neoplasms/genetics , Pathology, Molecular , Precision Medicine , High-Throughput Nucleotide Sequencing , Humans , Mutation , Neoplasms/therapy , Pilot Projects , Retrospective Studies , Switzerland
9.
Bioinformatics ; 34(1): 107-108, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28968639

ABSTRACT

Motivation: Next-generation sequencing is now an established method in genomics, and massive amounts of sequencing data are being generated on a regular basis. Analysis of the sequencing data is typically performed by lab-specific in-house solutions, but the agreement of results from different facilities is often small. General standards for quality control, reproducibility and documentation are missing. Results: We developed NGS-pipe, a flexible, transparent and easy-to-use framework for the design of pipelines to analyze whole-exome, whole-genome and transcriptome sequencing data. NGS-pipe facilitates the harmonization of genomic data analysis by supporting quality control, documentation, reproducibility, parallelization and easy adaptation to other NGS experiments. Availability and implementation: https://github.com/cbg-ethz/NGS-pipe. Contact: niko.beerenwinkel@bsse.ethz.ch.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods , Software , Gene Expression Profiling/methods , Gene Expression Profiling/standards , Genomics/methods , High-Throughput Nucleotide Sequencing/standards , Humans , Neoplasms/genetics , Reproducibility of Results , Sequence Analysis, DNA/standards , Sequence Analysis, RNA/standards
10.
BMC Bioinformatics ; 18(1): 8, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-28049408

ABSTRACT

BACKGROUND: Next-generation sequencing of matched tumor and normal biopsy pairs has become a technology of paramount importance for precision cancer treatment. Sequencing costs have dropped tremendously, allowing the sequencing of the whole exome of tumors for just a fraction of the total treatment costs. However, clinicians and scientists cannot take full advantage of the generated data because the accuracy of analysis pipelines is limited. This particularly concerns the reliable identification of subclonal mutations in a cancer tissue sample with very low frequencies, which may be clinically relevant. RESULTS: Using simulations based on kidney tumor data, we compared the performance of nine state-of-the-art variant callers, namely deepSNV, GATK HaplotypeCaller, GATK UnifiedGenotyper, JointSNVMix2, MuTect, SAMtools, SiNVICT, SomaticSniper, and VarScan2. The comparison was done as a function of variant allele frequencies and coverage. Our analysis revealed that deepSNV and JointSNVMix2 perform very well, especially in the low-frequency range. We attributed false positive and false negative calls of the nine tools to specific error sources and assigned them to processing steps of the pipeline. All of these errors can be expected to occur in real data sets. We found that modifying certain steps of the pipeline or parameters of the tools can lead to substantial improvements in performance. Furthermore, a novel integration strategy that combines the ranks of the variants yielded the best performance. More precisely, the rank-combination of deepSNV, JointSNVMix2, MuTect, SiNVICT and VarScan2 reached a sensitivity of 78% when fixing the precision at 90%, and outperformed all individual tools, where the maximum sensitivity was 71% with the same precision. CONCLUSIONS: The choice of well-performing tools for alignment and variant calling is crucial for the correct interpretation of exome sequencing data obtained from mixed samples, and common pipelines are suboptimal. We were able to relate observed substantial differences in performance to the underlying statistical models of the tools, and to pinpoint the error sources of false positive and false negative calls. These findings might inspire new software developments that improve exome sequencing pipelines and further the field of precision cancer treatment.


Subject(s)
Exome/genetics , Kidney Neoplasms/genetics , Algorithms , DNA, Neoplasm/chemistry , DNA, Neoplasm/metabolism , Genomics , High-Throughput Nucleotide Sequencing , Humans , Kidney Neoplasms/pathology , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
11.
J Mol Biol ; 428(1): 238-250, 2016 Jan 16.
Article in English | MEDLINE | ID: mdl-26711506

ABSTRACT

Determining the composition of viral populations is becoming increasingly important in the field of medical virology. While recently developed computational tools for viral haplotype analysis allow for correcting sequencing errors, they do not always allow for the removal of errors occurring in the upstream experimental protocol, such as PCR errors. Primer IDs (pIDs) are one method to address this problem by harnessing redundant template resampling for error correction. By using a reference mixture of five HIV-1 strains, we show how pIDs can be useful for estimating key experimental parameters, such as the substitution rate of the PCR process and the reverse transcription (RT) error rate. In addition, we introduce a hidden Markov model for determining the recombination rate of the RT PCR process. We found no strong sequence-specific bias in pID abundances (the same RT efficiencies as compared to commonly used short, specific RT primers) and no effects of pIDs on the estimated distribution of the references viruses.


Subject(s)
Genetic Variation , Genetics, Population/methods , HIV-1/classification , HIV-1/genetics , Molecular Biology/methods , DNA Primers/genetics , Quality Control
12.
Bioinformatics ; 30(17): i349-55, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25161219

ABSTRACT

MOTIVATION: Next-generation sequencing technologies produce unprecedented amounts of data, leading to completely new research fields. One of these is metagenomics, the study of large-size DNA samples containing a multitude of diverse organisms. A key problem in metagenomics is to functionally and taxonomically classify the sequenced DNA, to which end the well-known BLAST program is usually used. But BLAST has dramatic resource requirements at metagenomic scales of data, imposing a high financial or technical burden on the researcher. Multiple attempts have been made to overcome these limitations and present a viable alternative to BLAST. RESULTS: In this work we present Lambda, our own alternative for BLAST in the context of sequence classification. In our tests, Lambda often outperforms the best tools at reproducing BLAST's results and is the fastest compared with the current state of the art at comparable levels of sensitivity. AVAILABILITY AND IMPLEMENTATION: Lambda was implemented in the SeqAn open-source C++ library for sequence analysis and is publicly available for download at http://www.seqan.de/projects/lambda. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Metagenomics/methods , Sequence Alignment/methods , Sequence Analysis, DNA/methods , Algorithms , Software
13.
Article in English | MEDLINE | ID: mdl-20827403

ABSTRACT

The honeybee standard brain (HSB) serves as an interactive tool for relating morphologies of bee brain neurons and provides a reference system for functional and bibliographical properties (http://www.neurobiologie.fu-berlin.de/beebrain/). The ultimate goal is to document not only the morphological network properties of neurons collected from separate brains, but also to establish a graphical user interface for a neuron-related data base. Here, we review the current methods and protocols used to incorporate neuronal reconstructions into the HSB. Our registration protocol consists of two separate steps applied to imaging data from two-channel confocal microscopy scans: (1) The reconstruction of the neuron, facilitated by an automatic extraction of the neuron's skeleton based on threshold segmentation, and (2) the semi-automatic 3D segmentation of the neuropils and their registration with the HSB. The integration of neurons in the HSB is performed by applying the transformation computed in step (2) to the reconstructed neurons of step (1). The most critical issue of this protocol in terms of user interaction time - the segmentation process - is drastically improved by the use of a model-based segmentation process. Furthermore, the underlying statistical shape models (SSM) allow the visualization and analysis of characteristic variations in large sets of bee brain data. The anatomy of neural networks composed of multiple neurons that are registered into the HSB are visualized by depicting the 3D reconstructions together with semantic information with the objective to integrate data from multiple sources (electrophysiology, imaging, immunocytochemistry, molecular biology). Ultimately, this will allow the user to specify cell types and retrieve their morphologies along with physiological characterizations.

SELECTION OF CITATIONS
SEARCH DETAIL
...