scholarly journals SITH: an R package for visualizing and analyzing a spatial model of intratumor heterogeneity

2020 ◽  
Author(s):  
Phillip B. Nicol ◽  
Dániel L. Barabási ◽  
Amir Asiaee ◽  
Kevin R. Coombes

AbstractMotivationCancer progression, including the development of intratumor heterogeneity, is inherently a spatial process. Mathematical models of tumor evolution can provide insights into patterns of heterogeneity that can emerge in the presence of spatial growth.SummaryWe develop SITH, an R package that implements a lattice-based stochastic model of tumor growth and mutation. SITH provides 3D interactive visualizations of the simulated tumor and highlights heavily mutated regions. SITH can produce synthetic bulk and single-cell sequencing data sets by sampling from the tumor. The streamlined API will make SITH a useful tool for investigating the relationship between spatial growth and intratumor heterogeneity.Availability and ImplementationSITH is a part of CRAN and can thus be installed by running install.packages(“SITH”) from the R console. See https://CRAN.R-project.org/package=SITH for the user manual and package vignette.

2019 ◽  
Author(s):  
Navid Ahmadinejad ◽  
Shayna Troftgruben ◽  
Carlo Maley ◽  
Junwen Wang ◽  
Li Liu

ABSTRACTUnderstanding intratumor heterogeneity is critical to designing personalized treatments and improving clinical outcomes of cancers. Such investigations require accurate delineation of the subclonal composition of a tumor, which to date can only be reliably inferred from deep-sequencing data (>300x depth). To enable accurate subclonal discovery in tumors sequenced at standard depths (30-50x), we develop a novel computational method that incorporates an adaptive error model into statistical decomposition of mixed populations, which corrects the mean-variance dependency of sequencing data at the subclonal level. Tested on extensive computer simulations and real-world data, this new method, named model-based adaptive grouping of subclones (MAGOS), consistently outperforms existing methods on minimum sequencing depth, decomposition accuracy and computation efficiency. MAGOS supports subclone analysis using single nucleotide variants and copy number variants from one or more samples of an individual tumor. Applications of MAGOS to whole-exome sequencing data of 331 liver cancer samples discovered a significant association between subclonal diversity and patient overall survival. MAGOS is freely available as an R package at github (https://github.com/liliulab/magos).


2021 ◽  
Author(s):  
Farid Rashidi Mehrabadi ◽  
Kerrie L. Marie ◽  
Eva Perez-Guijarro ◽  
Salem Malikic ◽  
Erfan Sadeqi Azer ◽  
...  

Advances in single cell RNA sequencing (scRNAseq) technologies uncovered an unexpected complexity in solid tumors, underlining the relevance of intratumor heterogeneity for cancer progression and therapeutic resistance. Heterogeneity in the mutational composition of cancer cells is well captured by tumor phylogenies, which demonstrate how distinct cell populations evolve, and, e.g. develop metastatic potential or resistance to specific treatments. Unfortunately, because of their low read coverage per cell, mutation calls that can be made from scRNAseq data are very sparse and noisy. Additionally, available tumor phylogeny reconstruction methods cannot computationally handle a large number of cells and mutations present in typical scRNAseq datasets. Finally, there are no principled methods to assess distinct subclones observed in inferred tumor phylogenies and the genomic alterations that seed them. Here we present Trisicell, a computational toolkit for scalable tumor phylogeny reconstruction and evaluation from scRNAseq as well as single cell genome or exome sequencing data. Trisicell allows the identification of reliable subtrees of a tumor phylogeny, offering the ability to focus on the most important subclones and the genomic alterations that are associated with subclonal proliferation. We comprehensively assessed Trisicell on a melanoma model by comparing the phylogeny it builds using scRNAseq data, to those using matching bulk whole exome (bWES) and transcriptome (bWTS) sequencing data from clonal sublines derived from single cells. Our results demonstrate that tumor phylogenies based on mutation calls from scRNAseq data can be robustly inferred and evaluated by Trisicell. We also applied Trisicell to reconstruct and evaluate the phylogeny it builds using scRNAseq data from melanomas of the same mouse model after treatment with immune checkpoint blockade (ICB). After integratively analyzing our cell-specific mutation calls with their expression profiles, we observed that each subclone with a distinct set of novel somatic mutations is strongly associated with a distinct developmental status. Moreover, each subclone had developed a specific ICB-resistance mechanism. These results demonstrate that Trisicell can robustly utilize scRNAseq data to delineate intratumoral heterogeneity and tumor evolution.


BMC Genomics ◽  
2021 ◽  
Vol 22 (S5) ◽  
Author(s):  
Xikang Feng ◽  
Lingxi Chen ◽  
Yuhao Qing ◽  
Ruikang Li ◽  
Chaohui Li ◽  
...  

Abstract Background Copy number variation is crucial in deciphering the mechanism and cure of complex disorders and cancers. The recent advancement of scDNA sequencing technology sheds light upon addressing intratumor heterogeneity, detecting rare subclones, and reconstructing tumor evolution lineages at single-cell resolution. Nevertheless, the current circular binary segmentation based approach proves to fail to efficiently and effectively identify copy number shifts on some exceptional trails. Results Here, we propose SCYN, a CNV segmentation method powered with dynamic programming. SCYN resolves the precise segmentation on in silico dataset. Then we verified SCYN manifested accurate copy number inferring on triple negative breast cancer scDNA data, with array comparative genomic hybridization results of purified bulk samples as ground truth validation. We tested SCYN on two datasets of the newly emerged 10x Genomics CNV solution. SCYN successfully recognizes gastric cancer cells from 1% and 10% spike-ins 10x datasets. Moreover, SCYN is about 150 times faster than state of the art tool when dealing with the datasets of approximately 2000 cells. Conclusions SCYN robustly and efficiently detects segmentations and infers copy number profiles on single cell DNA sequencing data. It serves to reveal the tumor intra-heterogeneity. The source code of SCYN can be accessed in https://github.com/xikanfeng2/SCYN.


2021 ◽  
Vol 17 (12) ◽  
pp. e1009055
Author(s):  
Juan Diaz-Colunga ◽  
Ramon Diaz-Uriarte

Accurate prediction of tumor progression is key for adaptive therapy and precision medicine. Cancer progression models (CPMs) can be used to infer dependencies in mutation accumulation from cross-sectional data and provide predictions of tumor progression paths. However, their performance when predicting complete evolutionary trajectories is limited by violations of assumptions and the size of available data sets. Instead of predicting full tumor progression paths, here we focus on short-term predictions, more relevant for diagnostic and therapeutic purposes. We examine whether five distinct CPMs can be used to answer the question “Given that a genotype with n mutations has been observed, what genotype with n + 1 mutations is next in the path of tumor progression?” or, shortly, “What genotype comes next?”. Using simulated data we find that under specific combinations of genotype and fitness landscape characteristics CPMs can provide predictions of short-term evolution that closely match the true probabilities, and that some genotype characteristics can be much more relevant than global features. Application of these methods to 25 cancer data sets shows that their use is hampered by a lack of information needed to make principled decisions about method choice. Fruitful use of these methods for short-term predictions requires adapting method’s use to local genotype characteristics and obtaining reliable indicators of performance; it will also be necessary to clarify the interpretation of the method’s results when key assumptions do not hold.


2019 ◽  
Author(s):  
Haoyun Lei ◽  
Bochuan Lyu ◽  
E. Michael Gertz ◽  
Alejandro A. Schäffer ◽  
Xulian Shi ◽  
...  

AbstractCharacterizing intratumor heterogeneity (ITH) is crucial to understanding cancer development, but it is hampered by limits of available data sources. Bulk DNA sequencing is the most common technology to assess ITH, but mixes many genetically distinct cells in each sample, which must then be computationally deconvolved. Single-cell sequencing (SCS) is a promising alternative, but its limitations — e.g., high noise, difficulty scaling to large populations, technical artifacts, and large data sets — have so far made it impractical for studying cohorts of sufficient size to identify statistically robust features of tumor evolution. We have developed strategies for deconvolution and tumor phylogenetics combining limited amounts of bulk and single-cell data to gain some advantages of single-cell resolution with much lower cost, with specific focus on deconvolving genomic copy number data. We developed a mixed membership model for clonal deconvolution via non-negative matrix factorization (NMF) balancing deconvolution quality with similarity to single-cell samples via an associated efficient coordinate descent algorithm. We then improve on that algorithm by integrating deconvolution with clonal phylogeny inference, using a mixed integer linear programming (MILP) model to incorporate a minimum evolution phylogenetic tree cost in the problem objective. We demonstrate the effectiveness of these methods on semi-simulated data of known ground truth, showing improved deconvolution accuracy relative to bulk data alone.


2020 ◽  
Author(s):  
Xikang Feng ◽  
Lingxi Chen ◽  
Yuhao Qing ◽  
Ruikang Li ◽  
Chaohui Li ◽  
...  

Copy number variation is crucial in deciphering the mechanism and cure of complex disorders and cancers. The recent advancement of scDNA sequencing technology sheds light upon addressing intratumor heterogeneity, detecting rare subclones, and reconstructing tumor evolution lineages at single-cell resolution. Nevertheless, the current circular binary segmentation based approach proves to fail to efficiently and effectively identify copy number shifts on some exceptional trails. Here, we propose SCYN, a CNV segmentation method powered with dynamic programming. SCYN resolves the precise segmentation on two in silico datasets. Then we verified SCYN manifested accurate copy number inferring on triple negative breast cancer scDNA data, with array comparative genomic hybridization results of purified bulk samples as ground truth validation. We tested SCYN on two datasets of the newly emerged 10x Genomics CNV solution. SCYN successfully recognizes gastric cancer cells from 1% and 10% spike-ins 10x datasets. Moreover, SCYN is about 150 times faster than state of the art tool when dealing with the datasets of approximately 2000 cells. SCYN robustly and efficiently detects segmentations and infers copy number profiles on single cell DNA sequencing data. It serves to reveal the tumor intra-heterogeneity. The source code of SCYN can be accessed in https://github.com/xikanfeng2/SCYN. The visualization tools are hosted on https://sc.deepomics.org/.


2020 ◽  
Author(s):  
Juan Diaz-Colunga ◽  
Ramon Diaz-Uriarte

AbstractAccurate prediction of tumor progression is key for adaptive therapy and precision medicine. Cancer progression models (CPMs) can be used to infer dependencies in mutation accumulation from cross-sectional data and provide predictions of tumor progression paths. But their performance when predicting the complete evolutionary paths is limited by violations of assumptions and the size of available data sets. Instead of predicting full tumor progression paths, we can focus on short-term predictions, more relevant for diagnostic and therapeutic purposes. Here we examine if five distinct CPMs can be used to answer the question “Given that a genotype with n mutations has been observed, what genotype with n + 1 mutations is next in the path of tumor progression” or, shortly, “What genotype comes next”. Using simulated data we find that under specific combinations of genotype and fitness landscape characteristics CPMs can provide predictions of short-term evolution that closely match the true probabilities, and that some genotype characteristics (fitness and probability of being a local fitness maximum) can be much more relevant than global features. Thus, CPMs can provide short-term predictions even when global, long-term predictions are not possible because fitness landscape- and evolutionary model-specific assumptions are violated. When good performance is possible, we observe significant variation in the quality of predictions of different methods. Genotype-specific and global fitness landscape characteristics are required to determine which method provides best results in each case. Application of these methods to 25 cancer data sets shows that their use is hampered by lack of the information needed to make principled decisions about method choice and what predictions to trust. Fruitful use of these methods for short-term predictions requires adapting method’s use to local genotype characteristics and obtaining reliable indicators of performance; it will also be necessary to clarify the interpretation of the method’s results when key assumptions do not hold.


2015 ◽  
Author(s):  
Luca De Sano ◽  
Giulio Caravagna ◽  
Daniele Ramazzotti ◽  
Alex Graudenzi ◽  
Giancarlo Mauri ◽  
...  

AbstractMotivationWe introduce TRONCO (TRanslational ONCOlogy), an open-source R package that implements the state-of-the-art algorithms for the inference of cancer progression models from (epi)genomic mutational profiles. TRONCO can be used to extract population-level models describing the trends of accumulation of alterations in a cohort of cross-sectional samples, e.g., retrieved from publicly available databases, and individual-level models that reveal the clonal evolutionary history in single cancer patients, when multiple samples, e.g., multiple biopsies or single-cell sequencing data, are available. The resulting models can provide key hints in uncovering the evolutionary trajectories of cancer, especially for precision medicine or personalized therapy.AvailabilityTRONCO is released under the GPL license, it is hosted in the Software section at http://bimib.disco.unimib.it/ and archived also at [email protected]


2021 ◽  
Author(s):  
Xuan Cindy Li ◽  
Yuelin Liu ◽  
Farid Rashidi Mehrabadi ◽  
Salem Malikić ◽  
Stephen M. Mount ◽  
...  

AbstractRecent studies on the heritability of methylation patterns in tumor cells, suggest that tumor heterogeneity and progression can be studied through methylation changes. To elucidate methylation-based evolution trajectories in tumors, we introduce a novel computational frame-work for methylation phylogeny reconstruction, leveraging single cell bisulfite treated whole genome sequencing data (scBS-seq), additionally incorporating copy number information inferred independently from matched single cell RNA sequencing (scRNA-seq) data, when available. Our framework consists of three components: (i) noise-minimizing site selection, (ii) likelihood-based sequencing error correction, and (iii) pairwise expected distance calculation for cells, all designed to mitigate the effect of noise and uncertainty due to data sparsity commonly observed in scBS-seq data. We validate our approach with the scBS-seq data of multi-regionally sampled colorectal cancer cells, and demonstrate that the cell lineages constructed by our method strongly correlate with original sampling regions. Additionally, we show that the constructed phylogeny can be used to impute missing entries, which, in turn, may help reduce sparsity issues in scBS-seq data [email protected]


2019 ◽  
Author(s):  
Jessie Martin ◽  
Jason S. Tsukahara ◽  
Christopher Draheim ◽  
Zach Shipstead ◽  
Cody Mashburn ◽  
...  

**The uploaded manuscript is still in preparation** In this study, we tested the relationship between visual arrays tasks and working memory capacity and attention control. Specifically, we tested whether task design (selection or non-selection demands) impacted the relationship between visual arrays measures and constructs of working memory capacity and attention control. Using analyses from 4 independent data sets we showed that the degree to which visual arrays measures rely on selection influences the degree to which they reflect domain-general attention control.


Sign in / Sign up

Export Citation Format

Share Document