首页 > 最新文献

Computational and systems oncology最新文献

英文 中文
SITH: An R package for visualizing and analyzing a spatial model of intratumor heterogeneity 一个可视化和分析肿瘤内异质性空间模型的R包
Pub Date : 2022-05-31 DOI: 10.1002/cso2.1033
Phillip B. Nicol, Dániel L. Barabási, Kevin R. Coombes, Amir Asiaee

Cancer progression, including the development of intratumor heterogeneity, is inherently a spatial process. Mathematical models of tumor evolution may be a useful starting point for understanding the patterns of heterogeneity that can emerge in the presence of spatial growth. A commonly studied spatial growth model assumes that tumor cells occupy sites on a lattice and replicate into neighboring sites. Our R package SITH provides a convenient interface for exploring this model. Our efficient simulation algorithm allows for users to generate 3D tumors with millions of cells in under a minute. For the distribution of mutations throughout the tumor, SITH provides interactive graphics and summary plots. Additionally, SITH can produce synthetic bulk and single-cell DNA-seq datasets by sampling from the simulated tumor. A streamlined application programming interface (API) makes SITH a useful tool for investigating the relationship between spatial growth and intratumor heterogeneity. SITH is a part of CRAN and can 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.

癌症的进展,包括肿瘤内异质性的发展,本质上是一个空间过程。肿瘤进化的数学模型可能是理解在空间生长中可能出现的异质性模式的有用起点。一个普遍研究的空间生长模型假设肿瘤细胞占据晶格上的位置并复制到邻近的位置。我们的R包SITH为探索这个模型提供了一个方便的接口。我们高效的模拟算法允许用户在一分钟内生成具有数百万细胞的3D肿瘤。对于突变在整个肿瘤中的分布,SITH提供了交互式图形和汇总图。此外,通过从模拟肿瘤中取样,SITH可以生成合成的大块和单细胞DNA-seq数据集。简化的应用程序编程接口(API)使SITH成为研究空间生长和肿瘤内异质性之间关系的有用工具。SITH是CRAN的一部分,可以通过在R控制台中运行install.packages(“SITH”)来安装。请参阅https://CRAN.R-project.org/package=SITH获取用户手册和软件包说明。
{"title":"SITH: An R package for visualizing and analyzing a spatial model of intratumor heterogeneity","authors":"Phillip B. Nicol,&nbsp;Dániel L. Barabási,&nbsp;Kevin R. Coombes,&nbsp;Amir Asiaee","doi":"10.1002/cso2.1033","DOIUrl":"10.1002/cso2.1033","url":null,"abstract":"<p>Cancer progression, including the development of intratumor heterogeneity, is inherently a spatial process. Mathematical models of tumor evolution may be a useful starting point for understanding the patterns of heterogeneity that can emerge in the presence of spatial growth. A commonly studied spatial growth model assumes that tumor cells occupy sites on a lattice and replicate into neighboring sites. Our R package <i>SITH</i> provides a convenient interface for exploring this model. Our efficient simulation algorithm allows for users to generate 3D tumors with millions of cells in under a minute. For the distribution of mutations throughout the tumor, <i>SITH</i> provides interactive graphics and summary plots. Additionally, <i>SITH</i> can produce synthetic bulk and single-cell DNA-seq datasets by sampling from the simulated tumor. A streamlined application programming interface (API) makes <i>SITH</i> a useful tool for investigating the relationship between spatial growth and intratumor heterogeneity. <i>SITH</i> is a part of <span>CRAN</span> and can be installed by running <span>install.packages(“SITH”)</span> from the R console. See https://CRAN.R-project.org/package=SITH for the user manual and package vignette.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9374116/pdf/nihms-1801946.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9560891","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multiscale modeling of tumor adaption and invasion following anti-angiogenic therapy 抗血管生成治疗后肿瘤适应和侵袭的多尺度建模
Pub Date : 2022-02-21 DOI: 10.1002/cso2.1032
Colin G. Cess, Stacey D. Finley

In order to promote continued growth, a tumor must recruit new blood vessels, a process known as tumor angiogenesis. Many therapies have been tested that aim to inhibit tumor angiogenesis, with the goal of starving the tumor of nutrients and preventing tumor growth. However, many of these therapies have been unsuccessful and can paradoxically further tumor development by leading to increased local tumor invasion and metastasis. In this study, we use agent-based modeling to examine how hypoxic and acidic conditions following anti-angiogenic therapy can influence tumor development. Under these conditions, we find that cancer cells experience a phenotypic shift to a state of higher survival and invasive capability, spreading further away from the tumor into the surrounding tissue. Although anti-angiogenic therapy alone promotes tumor cell adaptation and invasiveness, we find that augmenting chemotherapy with anti-angiogenic therapy improves chemotherapeutic response and delays the time it takes for the tumor to regrow. Overall, we use computational modeling to explain the behavior of tumor cells in response to anti-angiogenic treatment in the dynamic tumor microenvironment.

为了促进肿瘤的持续生长,肿瘤必须招募新的血管,这一过程被称为肿瘤血管生成。已经测试了许多旨在抑制肿瘤血管生成的疗法,目的是使肿瘤缺乏营养,防止肿瘤生长。然而,许多这些治疗方法都不成功,并且可能通过导致局部肿瘤侵袭和转移增加而进一步发展肿瘤。在这项研究中,我们使用基于代理的模型来检查抗血管生成治疗后的缺氧和酸性条件如何影响肿瘤的发展。在这些条件下,我们发现癌细胞经历了表型转变,进入了更高存活率和侵袭能力的状态,从肿瘤向周围组织进一步扩散。虽然抗血管生成治疗单独促进肿瘤细胞的适应性和侵袭性,但我们发现抗血管生成治疗增加化疗可改善化疗反应并延迟肿瘤再生所需的时间。总的来说,我们使用计算模型来解释肿瘤细胞在动态肿瘤微环境中对抗血管生成治疗的反应行为。
{"title":"Multiscale modeling of tumor adaption and invasion following anti-angiogenic therapy","authors":"Colin G. Cess,&nbsp;Stacey D. Finley","doi":"10.1002/cso2.1032","DOIUrl":"10.1002/cso2.1032","url":null,"abstract":"<p>In order to promote continued growth, a tumor must recruit new blood vessels, a process known as tumor angiogenesis. Many therapies have been tested that aim to inhibit tumor angiogenesis, with the goal of starving the tumor of nutrients and preventing tumor growth. However, many of these therapies have been unsuccessful and can paradoxically further tumor development by leading to increased local tumor invasion and metastasis. In this study, we use agent-based modeling to examine how hypoxic and acidic conditions following anti-angiogenic therapy can influence tumor development. Under these conditions, we find that cancer cells experience a phenotypic shift to a state of higher survival and invasive capability, spreading further away from the tumor into the surrounding tissue. Although anti-angiogenic therapy alone promotes tumor cell adaptation and invasiveness, we find that augmenting chemotherapy with anti-angiogenic therapy improves chemotherapeutic response and delays the time it takes for the tumor to regrow. Overall, we use computational modeling to explain the behavior of tumor cells in response to anti-angiogenic treatment in the dynamic tumor microenvironment.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cso2.1032","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48554292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Multicellular mechanochemical hybrid cellular Potts model of tissue formation during epithelial-mesenchymal transition 上皮-间质转化过程中组织形成的多细胞机械化学杂交细胞Potts模型
Pub Date : 2021-12-27 DOI: 10.1002/cso2.1031
Shreyas U. Hirway, Christopher A. Lemmon, Seth H. Weinberg

Epithelial-mesenchymal transition (EMT) is the transdifferentiation of epithelial cells to a mesenchymal phenotype, in which cells lose epithelial-like cell–cell adhesions and gain mesenchymal-like enhanced contractility and mobility. EMT is crucial for tissue regeneration and is also implicated in pathological conditions, such as cancer metastasis. Prior work has shown that transforming growth factor-β1 (TGF-β1) is a potent inducer of this biological process. In this study, we develop a computational model coupling mechanical and biochemical signaling in a multicellular tissue undergoing EMT. Specifically, we utilize a recently developed formulation that integrates a multicellular cellular Potts model (CPM), a lattice-based stochastic model governing cell movement; a first moment of area model, governing cellular traction and junctional forces; a finite element model, which defines extracellular matrix (ECM) substrate strains; an intracellular signaling TGF-β1-mediated EMT model that governs cellular phenotype; and an extracellular signaling component governing ECM and TGF-β1 signaling. In this study, we modeled the spatial cellular patterns that occur in tissue and the ECM during EMT. Our model predicts that EMT often initially occurs at a tissue boundary due to mechanochemical coupling, which results in transdifferentiation to progress inwards toward the center. Variation in model parameters demonstrated conditions enhancing and suppressing EMT, especially to drive EMT in the absence of TGF-β1 and inhibit EMT in the presence of TGF-β1. Specifically, enhancing the mechanochemical feedback typically promoted EMT, whereas greater assembled ECM degradation suppressed EMT. Simulated scratch test experiments illustrate that ECM composition can impact closure directly through EMT signaling. In conclusion, we integrated mechanical, biochemical, and extracellular signaling networks in a novel hybrid computational model to reproduce tissue formation dynamics of EMT.

上皮-间充质转化(epithelial- mesenchymal transition, EMT)是上皮细胞向间充质表型的转分化,在此过程中,细胞失去上皮样细胞-细胞黏附,获得间充质样增强的收缩性和活动性。EMT对组织再生至关重要,也涉及病理条件,如癌症转移。先前的研究表明,转化生长因子- β1 (TGF- β1)是这一生物过程的有效诱导剂。在这项研究中,我们开发了一个计算模型耦合机械和生化信号在多细胞组织进行EMT。具体来说,我们利用最近开发的一种配方,该配方集成了多细胞细胞波茨模型(CPM),这是一种基于晶格的随机模型,用于控制细胞运动;区域模型的第一矩,控制细胞牵引力和连接力;定义细胞外基质(ECM)底物应变的有限元模型;调控细胞表型的细胞内信号传导TGF- β1介导的EMT模型;以及调控ECM和TGF- β1信号传导的细胞外信号成分。在这项研究中,我们模拟了EMT期间组织和ECM中发生的空间细胞模式。我们的模型预测,由于机械化学耦合,EMT通常最初发生在组织边界,导致转分化向中心进展。模型参数的变化显示了增强和抑制EMT的条件,特别是在TGF- β1不存在时驱动EMT,在TGF- β1存在时抑制EMT。具体来说,增强机械化学反馈通常会促进EMT,而更大的组装ECM降解会抑制EMT。模拟划痕测试实验表明,ECM成分可以通过EMT信号直接影响闭合。总之,我们将机械、生化和细胞外信号网络整合到一个新的混合计算模型中,以重现EMT的组织形成动力学。
{"title":"Multicellular mechanochemical hybrid cellular Potts model of tissue formation during epithelial-mesenchymal transition","authors":"Shreyas U. Hirway,&nbsp;Christopher A. Lemmon,&nbsp;Seth H. Weinberg","doi":"10.1002/cso2.1031","DOIUrl":"10.1002/cso2.1031","url":null,"abstract":"<p>Epithelial-mesenchymal transition (EMT) is the transdifferentiation of epithelial cells to a mesenchymal phenotype, in which cells lose epithelial-like cell–cell adhesions and gain mesenchymal-like enhanced contractility and mobility. EMT is crucial for tissue regeneration and is also implicated in pathological conditions, such as cancer metastasis. Prior work has shown that transforming growth factor-<math>\u0000 <mi>β</mi></math>1 (TGF-<math>\u0000 <mi>β</mi></math>1) is a potent inducer of this biological process. In this study, we develop a computational model coupling mechanical and biochemical signaling in a multicellular tissue undergoing EMT. Specifically, we utilize a recently developed formulation that integrates a multicellular cellular Potts model (CPM), a lattice-based stochastic model governing cell movement; a first moment of area model, governing cellular traction and junctional forces; a finite element model, which defines extracellular matrix (ECM) substrate strains; an intracellular signaling TGF-<math>\u0000 <mi>β</mi></math>1-mediated EMT model that governs cellular phenotype; and an extracellular signaling component governing ECM and TGF-<math>\u0000 <mi>β</mi></math>1 signaling. In this study, we modeled the spatial cellular patterns that occur in tissue and the ECM during EMT. Our model predicts that EMT often initially occurs at a tissue boundary due to mechanochemical coupling, which results in transdifferentiation to progress inwards toward the center. Variation in model parameters demonstrated conditions enhancing and suppressing EMT, especially to drive EMT in the absence of TGF-<math>\u0000 <mi>β</mi></math>1 and inhibit EMT in the presence of TGF-<math>\u0000 <mi>β</mi></math>1. Specifically, enhancing the mechanochemical feedback typically promoted EMT, whereas greater assembled ECM degradation suppressed EMT. Simulated scratch test experiments illustrate that ECM composition can impact closure directly through EMT signaling. In conclusion, we integrated mechanical, biochemical, and extracellular signaling networks in a novel hybrid computational model to reproduce tissue formation dynamics of EMT.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cso2.1031","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46509810","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 5
Dose-dependent mathematical modeling of interferon- α-treatment for personalized treatment of myeloproliferative neoplasms 干扰素- α-治疗骨髓增殖性肿瘤个体化治疗的剂量依赖性数学模型
Pub Date : 2021-12-07 DOI: 10.1002/cso2.1030
Rasmus K. Pedersen, Morten Andersen, Trine A. Knudsen, Vibe Skov, Lasse Kjær, Hans C. Hasselbalch, Johnny T. Ottesen

Long-term treatment with interferon-alfa (IFN) can reduce the disease burden of patients diagnosed with myeloproliferative neoplasms (MPNs). Determining individual patient responses to IFN therapy may allow for efficient personalized treatment, reducing both drop-out and disease burden. A mathematical model describing hematopoietic stem cells and the immune system is suggested. Considering the bone marrow and the blood allows for modeling disease dynamics both in the absence and presence of IFN treatment. Through comprehensive modeling of the effects of IFN, the model was related to individualized patient-data consisting of longitudinal hematologic and molecular measurements. Treatment responses were modeled on a population level, allowing for personalized predictions from a single pretreatment data point. Personalized fits were found to agree well with data for individual patients. This allowed for a quantitative description of the treatment response, yielding a mechanistic interpretation of differences from patient to patient. The treatment responses of individual patients were combined and a formulation of treatment responses on the population level was described and simulated. Based on pretreatment data and the actual treatment scheduling, the population-level response was found to predict the treatment response of particular patients accurately over a five-year period. Mechanism-based modeling of treatment effects demonstrates that hematologic and molecular observable quantities can be predicted on the level of individual patients. Personalized patient-fits suggest that the effect of IFN treatment can be quantified and interpreted through mathematical modeling, despite variation in hematologic and molecular responses between patients. Mathematical modeling suggests that in general both hematologic and molecular markers must be considered to avoid early relapse. Furthermore, personalized model-fits provide quantitative measures of the hematologic and molecular responses, determining when treatment-cessation is appropriate. Proof-of-concept population-level modeling of treatment responses from pretreatment data successfully predicted clinical measures for a 5-year period. We believe that this approach could have direct clinical relevance, offering expert guidance for clinical decisions about IFN treatment of MPN patients.

长期使用干扰素- α (IFN)治疗可减轻骨髓增生性肿瘤(mpn)患者的疾病负担。确定个体患者对干扰素治疗的反应可能允许有效的个性化治疗,减少退出和疾病负担。提出了描述造血干细胞和免疫系统的数学模型。考虑骨髓和血液允许在没有和存在干扰素治疗的情况下建立疾病动力学模型。通过对IFN效应的综合建模,该模型与个体化患者数据相关,包括纵向血液学和分子测量。治疗反应在人群水平上建模,允许从单个预处理数据点进行个性化预测。发现个性化契合度与个体患者的数据非常吻合。这允许对治疗反应进行定量描述,从而产生对患者之间差异的机制解释。将个体患者的治疗反应结合起来,并在人群水平上描述和模拟治疗反应的公式。根据预处理数据和实际的治疗计划,发现人群水平的反应可以准确地预测特定患者在五年期间的治疗反应。基于机制的治疗效果模型表明,血液学和分子观察量可以在个体患者的水平上进行预测。个性化的患者匹配表明,尽管患者之间的血液学和分子反应存在差异,但IFN治疗的效果可以通过数学模型进行量化和解释。数学模型表明,一般情况下,血液学和分子标记都必须考虑,以避免早期复发。此外,个性化模型拟合提供血液学和分子反应的定量测量,确定何时停止治疗是合适的。基于预处理数据的治疗反应的概念验证人群水平模型成功地预测了5年期间的临床措施。我们相信这种方法具有直接的临床意义,为临床决定IFN治疗MPN患者提供专家指导。
{"title":"Dose-dependent mathematical modeling of interferon-\u0000 α-treatment for personalized treatment of myeloproliferative neoplasms","authors":"Rasmus K. Pedersen,&nbsp;Morten Andersen,&nbsp;Trine A. Knudsen,&nbsp;Vibe Skov,&nbsp;Lasse Kjær,&nbsp;Hans C. Hasselbalch,&nbsp;Johnny T. Ottesen","doi":"10.1002/cso2.1030","DOIUrl":"10.1002/cso2.1030","url":null,"abstract":"<p>Long-term treatment with interferon-alfa (IFN) can reduce the disease burden of patients diagnosed with myeloproliferative neoplasms (MPNs). Determining individual patient responses to IFN therapy may allow for efficient personalized treatment, reducing both drop-out and disease burden. A mathematical model describing hematopoietic stem cells and the immune system is suggested. Considering the bone marrow and the blood allows for modeling disease dynamics both in the absence and presence of IFN treatment. Through comprehensive modeling of the effects of IFN, the model was related to individualized patient-data consisting of longitudinal hematologic and molecular measurements. Treatment responses were modeled on a population level, allowing for personalized predictions from a single pretreatment data point. Personalized fits were found to agree well with data for individual patients. This allowed for a quantitative description of the treatment response, yielding a mechanistic interpretation of differences from patient to patient. The treatment responses of individual patients were combined and a formulation of treatment responses on the population level was described and simulated. Based on pretreatment data and the actual treatment scheduling, the population-level response was found to predict the treatment response of particular patients accurately over a five-year period. Mechanism-based modeling of treatment effects demonstrates that hematologic and molecular observable quantities can be predicted on the level of individual patients. Personalized patient-fits suggest that the effect of IFN treatment can be quantified and interpreted through mathematical modeling, despite variation in hematologic and molecular responses between patients. Mathematical modeling suggests that in general both hematologic and molecular markers must be considered to avoid early relapse. Furthermore, personalized model-fits provide quantitative measures of the hematologic and molecular responses, determining when treatment-cessation is appropriate. Proof-of-concept population-level modeling of treatment responses from pretreatment data successfully predicted clinical measures for a 5-year period. We believe that this approach could have direct clinical relevance, offering expert guidance for clinical decisions about IFN treatment of MPN patients.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cso2.1030","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41806259","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Individual cell-based modeling of tumor cell plasticity-induced immune escape after CAR-T therapy CAR-T治疗后肿瘤细胞可塑性诱导免疫逃逸的个体细胞模型
Pub Date : 2021-09-23 DOI: 10.1002/cso2.1029
Can Zhang, Changrong Shao, Xiaopei Jiao, Yue Bai, Miao Li, Hanping Shi, Jinzhi Lei, Xiaosong Zhong

Chimeric antigen receptor (CAR) therapy targeting CD19 is an effective treatment for refractory B cell malignancies, especially B-cell acute lymphoblastic leukemia (B-ALL). The majority of patients achieve a complete response following a single infusion of CD19-targeted CAR-modified T cells (CAR-19 T cells); however, many patients suffer relapse after therapy, and the underlying mechanism remains unclear. To better understand the mechanism of tumor relapse, we developed an individual cell-based computational model based on major assumptions of the tumor cells heterogeneity and plasticity as well as the heterogeneous responses to CAR-T treatment. Model simulations reproduced the process of tumor relapse and predicted that cell plasticity induced by CAR-T stress can lead to tumor relapse in B-ALL. Model predictions were in agreement with experimental results of applying the second-generation CAR-T cells to mice injected with NALM-6-GL leukemic cells, in which 60% of the mice relapse within 3 months, relapsed tumors retained CD19 expression but exhibited a subpopulation of cells with high level CD34 transcription. The computational model suggests that the experimental data are compatible with a CAR-T cell-induced transition of tumor cells to hematopoietic stem-like cells and myeloid-like cells, which are resistant to the treatment. The proposed computational model framework was successfully developed to recapitulate the individual evolutionary dynamics and potentially allows to predict the outcomes of CAR-T treatment through model simulation based on early-stage observations of tumor burden and tumor cells analysis.

靶向CD19的嵌合抗原受体(CAR)治疗是治疗难治性B细胞恶性肿瘤,特别是B细胞急性淋巴细胞白血病(B- all)的有效方法。大多数患者在单次输注cd19靶向car修饰T细胞(CAR-19 T细胞)后获得完全缓解;然而,许多患者在治疗后复发,其潜在机制尚不清楚。为了更好地理解肿瘤复发的机制,我们基于肿瘤细胞的异质性和可塑性以及对CAR-T治疗的异质性反应的主要假设,开发了一个基于单个细胞的计算模型。模型模拟再现了肿瘤复发的过程,并预测CAR-T应激诱导的细胞可塑性可导致B-ALL肿瘤复发。模型预测与将第二代CAR-T细胞应用于注射了NALM-6-GL白血病细胞的小鼠的实验结果一致,其中60%的小鼠在3个月内复发,复发的肿瘤保留CD19表达,但表现出高水平CD34转录的细胞亚群。计算模型表明,实验数据与CAR-T细胞诱导的肿瘤细胞向造血干细胞样细胞和骨髓样细胞的转变是相容的,这些细胞对治疗有抵抗力。所提出的计算模型框架成功地概括了个体进化动力学,并有可能通过基于早期肿瘤负荷观察和肿瘤细胞分析的模型模拟来预测CAR-T治疗的结果。
{"title":"Individual cell-based modeling of tumor cell plasticity-induced immune escape after CAR-T therapy","authors":"Can Zhang,&nbsp;Changrong Shao,&nbsp;Xiaopei Jiao,&nbsp;Yue Bai,&nbsp;Miao Li,&nbsp;Hanping Shi,&nbsp;Jinzhi Lei,&nbsp;Xiaosong Zhong","doi":"10.1002/cso2.1029","DOIUrl":"10.1002/cso2.1029","url":null,"abstract":"<p>Chimeric antigen receptor (CAR) therapy targeting CD19 is an effective treatment for refractory B cell malignancies, especially B-cell acute lymphoblastic leukemia (B-ALL). The majority of patients achieve a complete response following a single infusion of CD19-targeted CAR-modified T cells (CAR-19 T cells); however, many patients suffer relapse after therapy, and the underlying mechanism remains unclear. To better understand the mechanism of tumor relapse, we developed an individual cell-based computational model based on major assumptions of the tumor cells heterogeneity and plasticity as well as the heterogeneous responses to CAR-T treatment. Model simulations reproduced the process of tumor relapse and predicted that cell plasticity induced by CAR-T stress can lead to tumor relapse in B-ALL. Model predictions were in agreement with experimental results of applying the second-generation CAR-T cells to mice injected with NALM-6-GL leukemic cells, in which 60% of the mice relapse within 3 months, relapsed tumors retained CD19 expression but exhibited a subpopulation of cells with high level CD34 transcription. The computational model suggests that the experimental data are compatible with a CAR-T cell-induced transition of tumor cells to hematopoietic stem-like cells and myeloid-like cells, which are resistant to the treatment. The proposed computational model framework was successfully developed to recapitulate the individual evolutionary dynamics and potentially allows to predict the outcomes of CAR-T treatment through model simulation based on early-stage observations of tumor burden and tumor cells analysis.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cso2.1029","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47154224","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Predicting time to relapse in acute myeloid leukemia through stochastic modeling of minimal residual disease based on clonality data 基于克隆数据的最小残留病随机模型预测急性髓系白血病复发时间
Pub Date : 2021-09-14 DOI: 10.1002/cso2.1026
Khanh N. Dinh, Roman Jaksik, Seth J. Corey, Marek Kimmel

Event-free and overall survival remain poor for patients with acute myeloid leukemia. Chemoresistant clones contributing to relapse arise from minimal residual disease (MRD) or newly acquired mutations. However, the dynamics of clones comprising MRD is poorly understood. We developed a predictive stochastic model, based on a multitype age-dependent Markov branching process, to describe how random events in MRD contribute to the heterogeneity in treatment response. We employed training and validation sets of patients who underwent whole-genome sequencing and for whom mutant clone frequencies at diagnosis and relapse were available. The disease evolution and treatment outcome are subject to stochastic fluctuations. Estimates of malignant clone growth rates, obtained by model fitting, are consistent with published data. Using the estimates from the training set, we developed a function linking MRD and time of relapse with MRD inferred from the model fits to clone frequencies and other data. An independent validation set confirmed our model. In a third dataset, we fitted the model to data at diagnosis and remission and predicted the time to relapse. As a conclusion, given bone marrow genome at diagnosis and MRD at or past remission, the model can predict time to relapse and help guide treatment decisions to mitigate relapse.

急性髓系白血病患者的无事件生存率和总生存率仍然很差。导致复发的耐药克隆来自微小残留病(MRD)或新获得的突变。然而,包括MRD的克隆的动力学知之甚少。我们建立了一个基于多类型年龄依赖的马尔可夫分支过程的预测随机模型,以描述MRD中的随机事件如何导致治疗反应的异质性。我们对接受全基因组测序的患者进行了培训和验证,并且在诊断和复发时可以获得突变克隆频率。疾病的发展和治疗结果受随机波动的影响。通过模型拟合获得的恶性克隆生长速率估计值与已发表的数据一致。使用来自训练集的估计,我们开发了一个函数,将MRD和复发时间与从模型拟合到克隆频率和其他数据推断的MRD联系起来。一个独立的验证集证实了我们的模型。在第三个数据集中,我们将模型拟合到诊断和缓解的数据中,并预测复发的时间。综上所述,根据诊断时的骨髓基因组和缓解时或过去的MRD,该模型可以预测复发时间,并帮助指导治疗决策以减轻复发。
{"title":"Predicting time to relapse in acute myeloid leukemia through stochastic modeling of minimal residual disease based on clonality data","authors":"Khanh N. Dinh,&nbsp;Roman Jaksik,&nbsp;Seth J. Corey,&nbsp;Marek Kimmel","doi":"10.1002/cso2.1026","DOIUrl":"10.1002/cso2.1026","url":null,"abstract":"<p>Event-free and overall survival remain poor for patients with acute myeloid leukemia. Chemoresistant clones contributing to relapse arise from minimal residual disease (MRD) or newly acquired mutations. However, the dynamics of clones comprising MRD is poorly understood. We developed a predictive stochastic model, based on a multitype age-dependent Markov branching process, to describe how random events in MRD contribute to the heterogeneity in treatment response. We employed training and validation sets of patients who underwent whole-genome sequencing and for whom mutant clone frequencies at diagnosis and relapse were available. The disease evolution and treatment outcome are subject to stochastic fluctuations. Estimates of malignant clone growth rates, obtained by model fitting, are consistent with published data. Using the estimates from the training set, we developed a function linking MRD and time of relapse with MRD inferred from the model fits to clone frequencies and other data. An independent validation set confirmed our model. In a third dataset, we fitted the model to data at diagnosis and remission and predicted the time to relapse. As a conclusion, given bone marrow genome at diagnosis and MRD at or past remission, the model can predict time to relapse and help guide treatment decisions to mitigate relapse.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1026","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39431799","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
A computational model for investigating the evolution of colonic crypts during Lynch syndrome carcinogenesis 研究Lynch综合征癌变过程中结肠隐窝进化的计算模型
Pub Date : 2021-07-04 DOI: 10.1002/cso2.1020
Saskia Haupt, Nils Gleim, Aysel Ahadova, Hendrik Bläker, Magnus von Knebel Doeberitz, Matthias Kloor, Vincent Heuveline

Lynch syndrome (LS), the most common inherited colorectal cancer (CRC) syndrome, increases the cancer risk in affected individuals. LS is caused by pathogenic germline variants in one of the DNA mismatch repair (MMR) genes, complete inactivation of which causes numerous mutations in affected cells. As CRC is believed to originate in colonic crypts, understanding the intra-crypt dynamics caused by mutational processes is essential for a complete picture of LS CRC and may have significant implications for cancer prevention.

We propose a computational model describing the evolution of colonic crypts during LS carcinogenesis. Extending existing modeling approaches for the non-Lynch scenario, we incorporated MMR deficiency and implemented recent experimental data demonstrating that somatic CTNNB1 mutations are common drivers of LS-associated CRCs, if affecting both alleles of the gene. Further, we simulated the effect of different mutations on the entire crypt, distinguishing non-transforming and transforming mutations.

As an example, we analyzed the spread of mutations in the genes APC and CTNNB1, which are frequently mutated in LS tumors, as well as of MMR deficiency itself. We quantified each mutation's potential for monoclonal conversion and investigated the influence of the cell location and of stem cell dynamics on mutation spread.

The in silico experiments underline the importance of stem cell dynamics for the overall crypt evolution. Further, simulating different mutational processes is essential in LS since mutations without survival advantages (the MMR deficiency-inducing second hit) play a key role. The effect of other mutations can be simulated with the proposed model. Our results provide first mathematical clues towards more effective surveillance protocols for LS carriers.

Lynch综合征(LS)是最常见的遗传性结直肠癌(CRC)综合征,它会增加受影响个体的癌症风险。LS是由一种DNA错配修复(MMR)基因的致病性种系变异引起的,该基因的完全失活导致受影响细胞发生大量突变。由于CRC被认为起源于结肠隐窝,了解突变过程引起的隐窝内动力学对于全面了解LS CRC至关重要,并可能对癌症预防具有重要意义。我们提出了一个计算模型来描述LS癌变过程中结肠隐窝的进化。将现有的建模方法扩展到非lynch情景,我们纳入了MMR缺陷,并实施了最近的实验数据,证明体细胞CTNNB1突变是ls相关crc的常见驱动因素,如果影响该基因的两个等位基因。此外,我们模拟了不同突变对整个隐窝的影响,区分了非转化突变和转化突变。作为一个例子,我们分析了在LS肿瘤中经常发生突变的基因APC和CTNNB1的突变传播,以及MMR缺陷本身。我们量化了每个突变的单克隆转化潜力,并研究了细胞位置和干细胞动力学对突变传播的影响。计算机实验强调了干细胞动力学对整个隐窝进化的重要性。此外,模拟不同的突变过程对LS至关重要,因为没有生存优势的突变(MMR缺陷诱导的二次命中)起着关键作用。其他突变的影响可以用所提出的模型来模拟。我们的结果为LS携带者更有效的监测协议提供了第一个数学线索。
{"title":"A computational model for investigating the evolution of colonic crypts during Lynch syndrome carcinogenesis","authors":"Saskia Haupt,&nbsp;Nils Gleim,&nbsp;Aysel Ahadova,&nbsp;Hendrik Bläker,&nbsp;Magnus von Knebel Doeberitz,&nbsp;Matthias Kloor,&nbsp;Vincent Heuveline","doi":"10.1002/cso2.1020","DOIUrl":"https://doi.org/10.1002/cso2.1020","url":null,"abstract":"<p>Lynch syndrome (LS), the most common inherited colorectal cancer (CRC) syndrome, increases the cancer risk in affected individuals. LS is caused by pathogenic germline variants in one of the DNA mismatch repair (MMR) genes, complete inactivation of which causes numerous mutations in affected cells. As CRC is believed to originate in colonic crypts, understanding the intra-crypt dynamics caused by mutational processes is essential for a complete picture of LS CRC and may have significant implications for cancer prevention.</p><p>We propose a computational model describing the evolution of colonic crypts during LS carcinogenesis. Extending existing modeling approaches for the non-Lynch scenario, we incorporated MMR deficiency and implemented recent experimental data demonstrating that somatic <i>CTNNB1</i> mutations are common drivers of LS-associated CRCs, if affecting both alleles of the gene. Further, we simulated the effect of different mutations on the entire crypt, distinguishing non-transforming and transforming mutations.</p><p>As an example, we analyzed the spread of mutations in the genes <i>APC</i> and <i>CTNNB1</i>, which are frequently mutated in LS tumors, as well as of MMR deficiency itself. We quantified each mutation's potential for monoclonal conversion and investigated the influence of the cell location and of stem cell dynamics on mutation spread.</p><p>The <i>in silico</i> experiments underline the importance of stem cell dynamics for the overall crypt evolution. Further, simulating different mutational processes is essential in LS since mutations without survival advantages (the MMR deficiency-inducing second hit) play a key role. The effect of other mutations can be simulated with the proposed model. Our results provide first mathematical clues towards more effective surveillance protocols for LS carriers.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1020","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"137795565","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of microRNAs in oncogenesis: Insights from computational and systems-level modeling approaches microrna在肿瘤发生中的作用:来自计算和系统级建模方法的见解
Pub Date : 2021-06-28 DOI: 10.1002/cso2.1028
Vinodhini Govindaraj, Sandip Kar

MicroRNAs (miRNAs) often govern the cell fate decision-making events associated with oncogenesis. miRNAs repress the target genes either by degrading the target mRNA or inhibiting the process of translation. However, mathematical and computational modeling of miRNA-mediated target gene regulation in various cellular network motifs indicates that miRNAs play a much more complex role in cellular decision-making events. In this review, we give an overview of the quantitative insights obtained from mathematical modeling of miRNA-mediated gene regulations by highlighting the various factors associated with it that are pivotal in diversifying the cell fate decisions related to oncogenesis. Intriguingly, recent experiments suggest that under certain circumstances, miRNAs can lead to more complex gene regulatory dynamics by causing target gene upregulation. We discuss these modeling approaches that can help in understanding the subtleties of miRNA effects in oncogenesis.

MicroRNAs (miRNAs)通常控制与肿瘤发生相关的细胞命运决策事件。mirna通过降解靶mRNA或抑制翻译过程来抑制靶基因。然而,各种细胞网络基序中mirna介导的靶基因调控的数学和计算模型表明,mirna在细胞决策事件中发挥的作用要复杂得多。在这篇综述中,我们概述了从mirna介导的基因调控的数学建模中获得的定量见解,强调了与之相关的各种因素,这些因素在与肿瘤发生相关的细胞命运决定的多样化中起着关键作用。有趣的是,最近的实验表明,在某些情况下,mirna可以通过引起靶基因上调而导致更复杂的基因调控动力学。我们讨论了这些建模方法,可以帮助理解miRNA在肿瘤发生中的微妙作用。
{"title":"Role of microRNAs in oncogenesis: Insights from computational and systems-level modeling approaches","authors":"Vinodhini Govindaraj,&nbsp;Sandip Kar","doi":"10.1002/cso2.1028","DOIUrl":"10.1002/cso2.1028","url":null,"abstract":"<p>MicroRNAs (miRNAs) often govern the cell fate decision-making events associated with oncogenesis. miRNAs repress the target genes either by degrading the target mRNA or inhibiting the process of translation. However, mathematical and computational modeling of miRNA-mediated target gene regulation in various cellular network motifs indicates that miRNAs play a much more complex role in cellular decision-making events. In this review, we give an overview of the quantitative insights obtained from mathematical modeling of miRNA-mediated gene regulations by highlighting the various factors associated with it that are pivotal in diversifying the cell fate decisions related to oncogenesis. Intriguingly, recent experiments suggest that under certain circumstances, miRNAs can lead to more complex gene regulatory dynamics by causing target gene upregulation. We discuss these modeling approaches that can help in understanding the subtleties of miRNA effects in oncogenesis.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1028","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41889484","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Oncogenetic network estimation with disjunctive Bayesian networks 基于析取贝叶斯网络的肿瘤发生网络估计
Pub Date : 2021-06-27 DOI: 10.1002/cso2.1027
Phillip B. Nicol, Kevin R. Coombes, Courtney Deaver, Oksana Chkrebtii, Subhadeep Paul, Amanda E. Toland, Amir Asiaee

Motivation: Cancer is the process of accumulating genetic alterations that confer selective advantages to tumor cells. The order in which aberrations occur is not arbitrary, and inferring the order of events is challenging due to the lack of longitudinal samples from tumors. Moreover, a network model of oncogenesis should capture biological facts such as distinct progression trajectories of cancer subtypes and patterns of mutual exclusivity of alterations in the same pathways.

In this paper, we present the disjunctive Bayesian network (DBN), a novel oncogenetic model with a phylogenetic interpretation. DBN is expressive enough to capture cancer subtypes' trajectories and mutually exclusive relations between alterations from unstratified data.

Results: In cases where the number of studied alterations is small (<30), we provide an efficient dynamic programming implementation of an exact structure learning method that finds a best DBN in the superexponential search space of networks. In rare cases that the number of alterations is large, we provided an efficient genetic algorithm in our software package, OncoBN. Through numerous synthetic and real data experiments, we show OncoBN's ability in inferring ground truth networks and recovering biologically meaningful progression networks.

Availability: OncoBN is implemented in R and is available at https://github.com/phillipnicol/OncoBN.

动机:癌症是一个积累遗传改变的过程,这些改变赋予肿瘤细胞选择性优势。畸变发生的顺序不是任意的,由于缺乏肿瘤的纵向样本,推断事件的顺序是具有挑战性的。此外,肿瘤发生的网络模型应该捕捉生物学事实,如癌症亚型的不同进展轨迹和相同途径中相互排他性改变的模式。在本文中,我们提出了析取贝叶斯网络(DBN),这是一种具有系统发育解释的新型肿瘤发生模型。DBN具有足够的表达能力,可以从非分层数据中捕捉癌症亚型的轨迹和变化之间的互斥关系。结果:在研究的改变数量较少的情况下(<30),我们提供了一种精确结构学习方法的高效动态规划实现,该方法在网络的超指数搜索空间中找到最佳DBN。在极少数情况下,如果改变的数量很大,我们在我们的软件包OncoBN中提供了一个有效的遗传算法。通过大量的合成和真实数据实验,我们证明了OncoBN在推断地面真值网络和恢复生物学上有意义的进展网络方面的能力。可用性:OncoBN是用R实现的,可以在https://github.com/phillipnicol/OncoBN上获得。
{"title":"Oncogenetic network estimation with disjunctive Bayesian networks","authors":"Phillip B. Nicol,&nbsp;Kevin R. Coombes,&nbsp;Courtney Deaver,&nbsp;Oksana Chkrebtii,&nbsp;Subhadeep Paul,&nbsp;Amanda E. Toland,&nbsp;Amir Asiaee","doi":"10.1002/cso2.1027","DOIUrl":"https://doi.org/10.1002/cso2.1027","url":null,"abstract":"<p><b>Motivation</b>: Cancer is the process of accumulating genetic alterations that confer selective advantages to tumor cells. The order in which aberrations occur is not arbitrary, and inferring the order of events is challenging due to the lack of longitudinal samples from tumors. Moreover, a network model of oncogenesis should capture biological facts such as distinct progression trajectories of cancer subtypes and patterns of mutual exclusivity of alterations in the same pathways.</p><p>In this paper, we present the disjunctive Bayesian network (DBN), a novel oncogenetic model with a phylogenetic interpretation. DBN is expressive enough to capture cancer subtypes' trajectories and mutually exclusive relations between alterations from unstratified data.</p><p><b>Results</b>: In cases where the number of studied alterations is small (<math>\u0000 <mrow>\u0000 <mo>&lt;</mo>\u0000 <mn>30</mn>\u0000 </mrow></math>), we provide an efficient dynamic programming implementation of an exact structure learning method that finds a best DBN in the superexponential search space of networks. In rare cases that the number of alterations is large, we provided an efficient genetic algorithm in our software package, OncoBN. Through numerous synthetic and real data experiments, we show OncoBN's ability in inferring ground truth networks and recovering biologically meaningful progression networks.</p><p><b>Availability</b>: OncoBN is implemented in R and is available at https://github.com/phillipnicol/OncoBN.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1027","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"137559155","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Computational systems-biology approaches for modeling gene networks driving epithelial–mesenchymal transitions 模拟驱动上皮-间质转化的基因网络的计算系统生物学方法
Pub Date : 2021-06-09 DOI: 10.1002/cso2.1021
Ataur Katebi, Daniel Ramirez, Mingyang Lu

Epithelial–mesenchymal transition (EMT) is an important biological process through which epithelial cells undergo phenotypic transitions to mesenchymal cells by losing cell–cell adhesion and gaining migratory properties that cells use in embryogenesis, wound healing, and cancer metastasis. An important research topic is to identify the underlying gene regulatory networks (GRNs) governing the decision making of EMT and develop predictive models based on the GRNs. The advent of recent genomic technology, such as single-cell RNA sequencing, has opened new opportunities to improve our understanding about the dynamical controls of EMT. In this article, we review three major types of computational and mathematical approaches and methods for inferring and modeling GRNs driving EMT. We emphasize (1) the bottom-up approaches, where GRNs are constructed through literature search; (2) the top-down approaches, where GRNs are derived from genome-wide sequencing data; (3) the combined top-down and bottom-up approaches, where EMT GRNs are constructed and simulated by integrating bioinformatics and mathematical modeling. We discuss the methodologies and applications of each approach and the available resources for these studies.

上皮-间充质转化(epithelial - mesenchymal transition, EMT)是一个重要的生物学过程,上皮细胞通过失去细胞间黏附和获得细胞在胚胎发生、伤口愈合和癌症转移中使用的迁移特性,经历表型转变为间充质细胞。一个重要的研究课题是识别控制EMT决策的潜在基因调控网络(grn),并基于grn建立预测模型。最近基因组技术的出现,如单细胞RNA测序,为提高我们对EMT动态控制的理解开辟了新的机会。在本文中,我们回顾了用于推断和建模驱动EMT的grn的三种主要类型的计算和数学方法。我们强调(1)自下而上的方法,通过文献检索构建grn;(2)自上而下的方法,其中grn来自全基因组测序数据;(3)自上而下和自下而上相结合的方法,将生物信息学和数学建模相结合,构建EMT grn并进行仿真。我们讨论了每种方法的方法和应用,以及这些研究的可用资源。
{"title":"Computational systems-biology approaches for modeling gene networks driving epithelial–mesenchymal transitions","authors":"Ataur Katebi,&nbsp;Daniel Ramirez,&nbsp;Mingyang Lu","doi":"10.1002/cso2.1021","DOIUrl":"10.1002/cso2.1021","url":null,"abstract":"<p>Epithelial–mesenchymal transition (EMT) is an important biological process through which epithelial cells undergo phenotypic transitions to mesenchymal cells by losing cell–cell adhesion and gaining migratory properties that cells use in embryogenesis, wound healing, and cancer metastasis. An important research topic is to identify the underlying gene regulatory networks (GRNs) governing the decision making of EMT and develop predictive models based on the GRNs. The advent of recent genomic technology, such as single-cell RNA sequencing, has opened new opportunities to improve our understanding about the dynamical controls of EMT. In this article, we review three major types of computational and mathematical approaches and methods for inferring and modeling GRNs driving EMT. We emphasize (1) the bottom-up approaches, where GRNs are constructed through literature search; (2) the top-down approaches, where GRNs are derived from genome-wide sequencing data; (3) the combined top-down and bottom-up approaches, where EMT GRNs are constructed and simulated by integrating bioinformatics and mathematical modeling. We discuss the methodologies and applications of each approach and the available resources for these studies.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-06-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1021","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39100629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 9
期刊
Computational and systems oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1