scholarly journals OncoGEMINI: Software for Investigating Tumor Variants From Multiple Biopsies With Integrated Cancer Annotations

Author(s):  
Thomas J. Nicholas ◽  
Michael J. Cormier ◽  
Xiaomeng Huang ◽  
Yi Qiao ◽  
Gabor T. Marth ◽  
...  

AbstractDNA sequencing has unveiled extensive tumor heterogeneity in several different cancer types, with many exhibiting diverse subclonal populations. Identifying and tracing mutations throughout the expansion and progression of a tumor represents a significant challenge. Furthermore, prioritizing the subset of such mutations most likely to contribute to tumor evolution or that could serve as potential therapeutic targets represents an ongoing problem. Here we describe OncoGEMINI, a new tool designed for exploring the complex patterns and trajectory of somatic and inherited variation observed in heterogeneous tumors biopsied over the course of treatment. This is accomplished by creating a searchable database of variants that includes tumor sampling timepoints and allows for filtering methods that reflect specific changes in variant allele frequencies over time. Additionally, by incorporating existing annotations and resources that facilitate the interpretation of cancer mutations (e.g., CIViC, DGIdb), OncoGEMINI enables rapid searches for, and potential identification of, mutations that may be driving subclonal evolution.

2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Thomas J. Nicholas ◽  
Michael J. Cormier ◽  
Xiaomeng Huang ◽  
Yi Qiao ◽  
Gabor T. Marth ◽  
...  

Abstract Background DNA sequencing has unveiled extensive tumor heterogeneity in several different cancer types, with many exhibiting diverse subclonal populations. Identifying and tracing mutations throughout the expansion and progression of a tumor represents a significant challenge. Furthermore, prioritizing the subset of such mutations most likely to contribute to tumor evolution or that could serve as potential therapeutic targets represents an ongoing problem. Results Here, we describe OncoGEMINI, a new tool designed for exploring the complex patterns and trajectory of somatic and inherited variation observed in heterogeneous tumors biopsied over the course of treatment. This is accomplished by creating a searchable database of variants that includes tumor sampling time points and allows for filtering methods that reflect specific changes in variant allele frequencies over time. Additionally, by incorporating existing annotations and resources that facilitate the interpretation of cancer mutations (e.g., CIViC, DGIdb), OncoGEMINI enables rapid searches for, and potential identification of, mutations that may be driving subclonal evolution. Conclusions By combining relevant genomic annotations alongside specific filtering tools, OncoGEMINI provides powerful and customizable approaches that enable the quick identification of individual tumor variants that meet specified criteria. It can be applied to a wide range of tumor-derived sequence data, but is especially designed for studies with multiple samples, including longitudinal datasets. It is available under an MIT license at github.com/fakedrtom/oncogemini.


Cancers ◽  
2021 ◽  
Vol 13 (8) ◽  
pp. 1800
Author(s):  
Giusi Russo ◽  
Alfonso Tramontano ◽  
Ilaria Iodice ◽  
Lorenzo Chiariotti ◽  
Antonio Pezone

Cancer evolution is associated with genomic instability and epigenetic alterations, which contribute to the inter and intra tumor heterogeneity, making genetic markers not accurate to monitor tumor evolution. Epigenetic changes, aberrant DNA methylation and modifications of chromatin proteins, determine the “epigenome chaos”, which means that the changes of epigenetic traits are randomly generated, but strongly selected by deterministic events. Disordered changes of DNA methylation profiles are the hallmarks of all cancer types, but it is not clear if aberrant methylation is the cause or the consequence of cancer evolution. Critical points to address are the profound epigenetic intra- and inter-tumor heterogeneity and the nature of the heterogeneity of the methylation patterns in each single cell in the tumor population. To analyze the methylation heterogeneity of tumors, new technological and informatic tools have been developed. This review discusses the state of the art of DNA methylation analysis and new approaches to reduce or solve the complexity of methylated alleles in DNA or cell populations.


2018 ◽  
Author(s):  
Stefan C. Dentro ◽  
Ignaty Leshchiner ◽  
Kerstin Haase ◽  
Maxime Tarabichi ◽  
Jeff Wintersinger ◽  
...  

SUMMARYIntra-tumor heterogeneity (ITH) is a mechanism of therapeutic resistance and therefore an important clinical challenge. However, the extent, origin and drivers of ITH across cancer types are poorly understood. To address this question, we extensively characterize ITH across whole-genome sequences of 2,658 cancer samples, spanning 38 cancer types. Nearly all informative samples (95.1%) contain evidence of distinct subclonal expansions, with frequent branching relationships between subclones. We observe positive selection of subclonal driver mutations across most cancer types, and identify cancer type specific subclonal patterns of driver gene mutations, fusions, structural variants and copy-number alterations, as well as dynamic changes in mutational processes between subclonal expansions. Our results underline the importance of ITH and its drivers in tumor evolution, and provide an unprecedented pan-cancer resource of comprehensively annotated subclonal events from whole-genome sequencing data.


2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Leah L. Weber ◽  
Mohammed El-Kebir

Abstract Background Cancer arises from an evolutionary process where somatic mutations give rise to clonal expansions. Reconstructing this evolutionary process is useful for treatment decision-making as well as understanding evolutionary patterns across patients and cancer types. In particular, classifying a tumor’s evolutionary process as either linear or branched and understanding what cancer types and which patients have each of these trajectories could provide useful insights for both clinicians and researchers. While comprehensive cancer phylogeny inference from single-cell DNA sequencing data is challenging due to limitations with current sequencing technology and the complexity of the resulting problem, current data might provide sufficient signal to accurately classify a tumor’s evolutionary history as either linear or branched. Results We introduce the Linear Perfect Phylogeny Flipping (LPPF) problem as a means of testing two alternative hypotheses for the pattern of evolution, which we prove to be NP-hard. We develop Phyolin, which uses constraint programming to solve the LPPF problem. Through both in silico experiments and real data application, we demonstrate the performance of our method, outperforming a competing machine learning approach. Conclusion Phyolin is an accurate, easy to use and fast method for classifying an evolutionary trajectory as linear or branched given a tumor’s single-cell DNA sequencing data.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A18-A18
Author(s):  
Jaeyoun Choi ◽  
Myungwoo Nam ◽  
Stanislav Fridland ◽  
Jinyoung Hwang ◽  
Chan Mi Jung ◽  
...  

BackgroundTumor heterogeneity assessment may help predict response to immunotherapy. In melanoma mouse models, tumor heterogeneity impaired immune response.1 In addition, among lung cancer patients receiving immunotherapy, the high clonal neoantigen group had favorable survival and outcomes.2 Ideal methods of quantifying tumor heterogeneity are multiple biopsies or autopsy. However, these are not feasible in routine clinical practice. Circulating tumor DNA (ctDNA) is emerging as an alternative. Here, we reviewed the current state of tumor heterogeneity quantification from ctDNA. Furthermore, we propose a new tumor heterogeneity index(THI) based on our own scoring system, utilizing both ctDNA and tissue DNA.MethodsSystematic literature search on Pubmed was conducted up to August 18, 2020. A scoring system and THI were theoretically derived.ResultsTwo studies suggested their own methods of assessing tumor heterogeneity. One suggested clustering mutations with Pyclone,3 and the other suggested using the ratio of allele frequency (AF) to the maximum somatic allele frequency (MSAF).4 According to the former, the mutations in the highest cellular prevalence cluster can be defined as clonal mutations. According to the latter, the mutations with AF/MSAF<10% can be defined as subclonal mutations. To date, there have been no studies on utilizing both ctDNA and tissue DNA simultaneously to quantify tumor heterogeneity. We hypothesize that a mutation found in only one of either ctDNA or tissue DNA has a higher chance of being subclonal.We suggest a scoring system based on the previously mentioned methods to estimate the probability for a mutant allele to be subclonal. Adding up the points that correspond to the conditions results in a subclonality score (table 1). In a given ctDNA, the number of alleles with a subclonality score greater than or equal to 2 divided by the total number of alleles is defined as blood THI (bTHI) (figure 1). We can repeat the same calculation in a given tissue DNA for tissue THI (tTHI) (figure 2). Finally, we define composite THI (cTHI) as the mean of bTHI and tTHI.Abstract 18 Table 1Subclonality scoreAbstract 18 Figure 1Hypothetical distribution of all alleles found in ctDNA bTHI = the number of alleles with a subclonality score greater than or equal to 2/the total number of alleles found in ctDNA = 10/20 =50%Abstract 18 Figure 2Hypothetical distribution of all alleles found in tissue DNA tTHI= the number of alleles with a subclonality score greater than or equal to 2/the total number of alleles found in tissue DNA = 16/40 = 40% cTHI= (bTHI + tTHI)/2 = 45%ConclusionsTumor heterogeneity is becoming an important biomarker for predicting response to immunotherapy. Because autopsy and multiple biopsies are not feasible, utilizing both ctDNA and tissue DNA is the most comprehensive and practical approach. Therefore, we propose cTHI, for the first time, as a quantification measure of tumor heterogeneity.ReferencesWolf Y, Bartok O. UVB-Induced Tumor Heterogeneity Diminishes Immune Response in Melanoma. Cell 2019;179:219–235.McGranahan N, Swanton C. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351:1463–1469.Ma F, Guan Y. Assessing tumor heterogeneity using ctDNA to predict and monitor therapeutic response in metastatic breast cancer. Int J Cancer 2020;146:1359–1368.Liu Z, Xie Z. Presence of allele frequency heterogeneity defined by ctDNA profiling predicts unfavorable overall survival of NSCLC. Transl Lung Cancer Res 2019;8:1045–1050.


2021 ◽  
Vol 17 (3) ◽  
pp. 1-38
Author(s):  
Lauren Biernacki ◽  
Mark Gallagher ◽  
Zhixing Xu ◽  
Misiker Tadesse Aga ◽  
Austin Harris ◽  
...  

There is an increasing body of work in the area of hardware defenses for software-driven security attacks. A significant challenge in developing these defenses is that the space of security vulnerabilities and exploits is large and not fully understood. This results in specific point defenses that aim to patch particular vulnerabilities. While these defenses are valuable, they are often blindsided by fresh attacks that exploit new vulnerabilities. This article aims to address this issue by suggesting ways to make future defenses more durable based on an organization of security vulnerabilities as they arise throughout the program life cycle. We classify these vulnerability sources through programming, compilation, and hardware realization, and we show how each source introduces unintended states and transitions into the implementation. Further, we show how security exploits gain control by moving the implementation to an unintended state using knowledge of these sources and how defenses work to prevent these transitions. This framework of analyzing vulnerability sources, exploits, and defenses provides insights into developing durable defenses that could defend against broader categories of exploits. We present illustrative case studies of four important attack genealogies—showing how they fit into the presented framework and how the sophistication of the exploits and defenses have evolved over time, providing us insights for the future.


2018 ◽  
Vol 115 (47) ◽  
pp. E11101-E11110 ◽  
Author(s):  
Erez Persi ◽  
Yuri I. Wolf ◽  
Mark D. M. Leiserson ◽  
Eugene V. Koonin ◽  
Eytan Ruppin

How mutation and selection determine the fitness landscape of tumors and hence clinical outcome is an open fundamental question in cancer biology, crucial for the assessment of therapeutic strategies and resistance to treatment. Here we explore the mutation-selection phase diagram of 6,721 tumors representing 23 cancer types by quantifying the overall somatic point mutation load (ML) and selection (dN/dS) in the entire proteome of each tumor. We show that ML strongly correlates with patient survival, revealing two opposing regimes around a critical point. In low-ML cancers, a high number of mutations indicates poor prognosis, whereas high-ML cancers show the opposite trend, presumably due to mutational meltdown. Although the majority of cancers evolve near neutrality, deviations are observed at extreme MLs. Melanoma, with the highest ML, evolves under purifying selection, whereas in low-ML cancers, signatures of positive selection are observed, demonstrating how selection affects tumor fitness. Moreover, different cancers occupy specific positions on the ML–dN/dS plane, revealing a diversity of evolutionary trajectories. These results support and expand the theory of tumor evolution and its nonlinear effects on survival.


Cancers ◽  
2021 ◽  
Vol 13 (18) ◽  
pp. 4560
Author(s):  
Jerome Griffon ◽  
Delphine Buffello ◽  
Alain Giron ◽  
S. Lori Bridal ◽  
Michele Lamuraglia

Purpose: There is a clinical need to better non-invasively characterize the tumor microenvironment in order to reveal evidence of early tumor response to therapy and to better understand therapeutic response. The goals of this work are first to compare the sensitivity to modifications occurring during tumor growth for measurements of tumor volume, immunohistochemistry parameters, and emerging ultrasound parameters (Shear Wave Elastography (SWE) and dynamic Contrast-Enhanced Ultrasound (CEUS)), and secondly, to study the link between the different parameters. Methods: Five different groups of 9 to 10 BALB/c female mice with subcutaneous CT26 tumors were imaged using B-mode morphological imaging, SWE, and CEUS at different dates. Whole-slice immunohistological data stained for the nuclei, T lymphocytes, apoptosis, and vascular endothelium from these tumors were analyzed. Results: Tumor volume and three CEUS parameters (Time to Peak, Wash-In Rate, and Wash-Out Rate) significantly changed over time. The immunohistological parameters, CEUS parameters, and SWE parameters showed intracorrelation. Four immunohistological parameters (the number of T lymphocytes per mm2 and its standard deviation, the percentage area of apoptosis, and the colocalization of apoptosis and vascular endothelium) were correlated with the CEUS parameters (Time to Peak, Wash-In Rate, Wash-Out Rate, and Mean Transit Time). The SWE parameters were not correlated with the CEUS parameters nor with the immunohistological parameters. Conclusions: US imaging can provide additional information on tumoral changes. This could help to better explore the effect of therapies on tumor evolution, by studying the evolution of the parameters over time and by studying their correlations.


2018 ◽  
Author(s):  
An-Shun Tai ◽  
Chien-Hua Peng ◽  
Shih-Chi Peng ◽  
Wen-Ping Hsieh

AbstractMultistage tumorigenesis is a dynamic process characterized by the accumulation of mutations. Thus, a tumor mass is composed of genetically divergent cell subclones. With the advancement of next-generation sequencing (NGS), mathematical models have been recently developed to decompose tumor subclonal architecture from a collective genome sequencing data. Most of the methods focused on single-nucleotide variants (SNVs). However, somatic copy number aberrations (CNAs) also play critical roles in carcinogenesis. Therefore, further modeling subclonal CNAs composition would hold the promise to improve the analysis of tumor heterogeneity and cancer evolution. To address this issue, we developed a two-way mixture Poisson model, named CloneDeMix for the deconvolution of read-depth information. It can infer the subclonal copy number, mutational cellular prevalence (MCP), subclone composition, and the order in which mutations occurred in the evolutionary hierarchy. The performance of CloneDeMix was systematically assessed in simulations. As a result, the accuracy of CNA inference was nearly 93% and the MCP was also accurately restored. Furthermore, we also demonstrated its applicability using head and neck cancer samples from TCGA. Our results inform about the extent of subclonal CNA diversity, and a group of candidate genes that probably initiate lymph node metastasis during tumor evolution was also discovered. Most importantly, these driver genes are located at 11q13.3 which is highly susceptible to copy number change in head and neck cancer genomes. This study successfully estimates subclonal CNAs and exhibit the evolutionary relationships of mutation events. By doing so, we can track tumor heterogeneity and identify crucial mutations during evolution process. Hence, it facilitates not only understanding the cancer development but finding potential therapeutic targets. Briefly, this framework has implications for improved modeling of tumor evolution and the importance of inclusion of subclonal CNAs.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-37-SCI-37
Author(s):  
Christina Curtis

Abstract Cancer results from the acquisition of somatic alterations in an evolutionary process that typically occurs over many years, much of which is occult. Understanding the evolutionary dynamics that are operative at different stages of progression in individual tumors might inform the earlier detection, diagnosis, and treatment of cancer. For decades, tumor progression has been described as a gradual stepwise process, and it is through this lens that the underlying mechanisms have been interpreted and therapeutic strategies have been developed. Although these processes cannot be directly observed, the resultant spatiotemporal patterns of genetic variation amongst tumor cells encode their evolutionary histories. Cancer genome sequencing has thus yielded unprecedented insights into intra-tumor heterogeneity (ITH) and these data enable the inference of tumor dynamics using population genetics techniques. The application of such approaches suggests that tumor evolution is not necessarily gradual, but rather can be punctuated, resulting in revision of the de facto sequential clonal expansion model. For example, we previously described a Big Bang model of human colorectal tumor growth, wherein after transformation the neoplasm grows predominantly as a single terminal expansion in the absence of stringent selection, compatible with effectively neutral evolution1. In the Big Bang model, the timing of a mutation is the fundamental determinant of its frequency in the final tumor such that all major clones persist during growth and most detectable intra-tumor heterogeneity (ITH) occurs early. By analyzing multi-region and single gland genomic profiles in colorectal adenomas and carcinomas within a spatial agent-based tumor growth model and Bayesian statistical inference framework, we demonstrated the early origin of ITH and verified several other predictions of the Big Bang model. This new model provides a quantitative framework for understanding tumor progression with several clinical implications. In particular, rare but potentially aggressive subclones may be undetectable, providing a rich substrate for the emergence of resistance under treatment selective pressure. These data also suggest that some tumors may be born to be bad, wherein malignant potential is specified early. While not all tumors exhibit Big Bang dynamics, effectively neutral evolution has since been reported in other tumors and hence may be relatively common. These findings emphasize the need for methods to infer the role of selection in established human tumors and the systematic evaluation of distinct modes of evolution across tumor types and disease stages. To address this need, we developed an extensible population genetics framework to simulate spatial tumor growth and evaluate evidence for different evolutionary modes based on patterns of genetic variation derived from multi-region sequencing (MRS) data2. We demonstrate that while it is feasible to distinguish strong positive selection from neutral tumor evolution, weak selection and neutral evolution were indistinguishable in current data. Building on these findings, we developed a classifier that exploits novel measures of ITH and applied this to MRS data from diverse tumor types, revealing different evolutionary modes amongst treatment naïve tumors. To better understand evolutionary tempos during disease progression, we further characterized longitudinally sampled specimens. These findings have implications for forecasting tumor evolution and designing more effective treatment strategies. 1. Sottoriva A, Kang H, Ma Z, et al. A Big Bang model of human colorectal tumor growth. Nature Genetics. 2015;47:209-16. 2. Sun R, Hu Z, Sottoriva A, et al. Between-region genetic divergence reflects the mode and tempo of tumor evolution. Nature Genetics. 2017;49:1015-24. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document