首页 > 最新文献

Computational and systems oncology最新文献

英文 中文
A validated mathematical model of FGFR3-mediated tumor growth reveals pathways to harness the benefits of combination targeted therapy and immunotherapy in bladder cancer 一个经过验证的fgfr3介导肿瘤生长的数学模型揭示了利用联合靶向治疗和免疫治疗在膀胱癌中的益处的途径
Pub Date : 2021-05-19 DOI: 10.1002/cso2.1019
Kamaldeen Okuneye, Daniel Bergman, Jeffrey C. Bloodworth, Alexander T. Pearson, Randy F. Sweis, Trachette L. Jackson

Bladder cancer is a common malignancy with over 80,000 estimated new cases and nearly 18,000 deaths per year in the United States alone. Therapeutic options for metastatic bladder cancer had not evolved much for nearly four decades, until recently, when five immune checkpoint inhibitors were approved by the U.S. Food and Drug Administration (FDA). Despite the activity of these drugs in some patients, the objective response rate for each is less than 25%. At the same time, fibroblast growth factor receptors (FGFRs) have been attractive drug targets for a variety of cancers, and in 2019 the FDA approved the first therapy targeted against FGFR3 for bladder cancer. Given the excitement around these new receptor tyrosine kinase and immune checkpoint targeted strategies, and the challenges they each may face on their own, emerging data suggest that combining these treatment options could lead to improved therapeutic outcomes. In this paper, we develop a mathematical model for FGFR3-mediated tumor growth and use it to investigate the impact of the combined administration of a small molecule inhibitor of FGFR3 and a monoclonal antibody against the PD-1/PD-L1 immune checkpoint. The model is carefully calibrated and validated with experimental data before survival benefits, and dosing schedules are explored. Predictions of the model suggest that FGFR3 mutation reduces the effectiveness of anti-PD-L1 therapy, that there are regions of parameter space where each monotherapy can outperform the other, and that pretreatment with anti-PD-L1 therapy always results in greater tumor reduction even when anti-FGFR3 therapy is the more effective monotherapy.

膀胱癌是一种常见的恶性肿瘤,据估计,仅在美国,每年就有超过80,000例新病例和近18,000例死亡。近四十年来,转移性膀胱癌的治疗选择并没有太大的发展,直到最近,美国食品和药物管理局(FDA)批准了五种免疫检查点抑制剂。尽管这些药物在一些患者中有活性,但每种药物的客观缓解率都低于25%。与此同时,成纤维细胞生长因子受体(FGFRs)已成为多种癌症的有吸引力的药物靶点,2019年FDA批准了首个针对FGFR3治疗膀胱癌的药物。鉴于对这些新的受体酪氨酸激酶和免疫检查点靶向策略的兴奋,以及它们各自可能面临的挑战,新出现的数据表明,结合这些治疗方案可能会改善治疗结果。在本文中,我们建立了FGFR3介导的肿瘤生长的数学模型,并用它来研究FGFR3的小分子抑制剂和针对PD-1/PD-L1免疫检查点的单克隆抗体联合施用的影响。在获得生存效益之前,该模型经过仔细校准和实验数据验证,并探索了给药方案。该模型的预测表明,FGFR3突变降低了抗pd - l1治疗的有效性,存在参数空间区域,每种单一疗法都可以优于其他疗法,并且抗pd - l1治疗的预处理总是导致更大的肿瘤缩小,即使抗FGFR3治疗是更有效的单一疗法。
{"title":"A validated mathematical model of FGFR3-mediated tumor growth reveals pathways to harness the benefits of combination targeted therapy and immunotherapy in bladder cancer","authors":"Kamaldeen Okuneye,&nbsp;Daniel Bergman,&nbsp;Jeffrey C. Bloodworth,&nbsp;Alexander T. Pearson,&nbsp;Randy F. Sweis,&nbsp;Trachette L. Jackson","doi":"10.1002/cso2.1019","DOIUrl":"10.1002/cso2.1019","url":null,"abstract":"<p>Bladder cancer is a common malignancy with over 80,000 estimated new cases and nearly 18,000 deaths per year in the United States alone. Therapeutic options for metastatic bladder cancer had not evolved much for nearly four decades, until recently, when five immune checkpoint inhibitors were approved by the U.S. Food and Drug Administration (FDA). Despite the activity of these drugs in some patients, the objective response rate for each is less than 25%. At the same time, fibroblast growth factor receptors (FGFRs) have been attractive drug targets for a variety of cancers, and in 2019 the FDA approved the first therapy targeted against FGFR3 for bladder cancer. Given the excitement around these new receptor tyrosine kinase and immune checkpoint targeted strategies, and the challenges they each may face on their own, emerging data suggest that combining these treatment options could lead to improved therapeutic outcomes. In this paper, we develop a mathematical model for FGFR3-mediated tumor growth and use it to investigate the impact of the combined administration of a small molecule inhibitor of FGFR3 and a monoclonal antibody against the PD-1/PD-L1 immune checkpoint. The model is carefully calibrated and validated with experimental data before survival benefits, and dosing schedules are explored. Predictions of the model suggest that FGFR3 mutation reduces the effectiveness of anti-PD-L1 therapy, that there are regions of parameter space where each monotherapy can outperform the other, and that pretreatment with anti-PD-L1 therapy always results in greater tumor reduction even when anti-FGFR3 therapy is the more effective monotherapy.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1019","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39898438","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: 6
Biomechanical modelling of cancer: Agent-based force-based models of solid tumours within the context of the tumour microenvironment 癌症的生物力学建模:肿瘤微环境下实体肿瘤的基于agent的力模型
Pub Date : 2021-05-18 DOI: 10.1002/cso2.1018
Cicely K. Macnamara

Once cancer is initiated, with normal cells mutated into malignant ones, a solid tumour grows, develops and spreads within its microenvironment invading the local tissue; the disease progresses and the cancer cells migrate around the body leading to metastasis, the formation of distant secondary tumours. Interactions between the tumour and its microenvironment drive this cascade of events which have devastating, if not fatal, consequences for the human host/patient. Among these interactions, biomechanical interactions are a vital component. In this review paper, key biomechanical relationships are discussed through a presentation of modelling efforts by the mathematical and computational oncology community. The main focus is directed, naturally, towards lattice-free agent-based, force-based models of solid tumour growth and development. In such models, interactions between pairs of cancer cells (as well as between cells and other structures of the tumour microenvironment) are governed by forces. These forces are ones of repulsion and adhesion, and are typically modelled via either an extended Hertz model of contact mechanics or using Johnson–Kendal–Roberts theory, both of which are discussed here. The role of the extracellular matrix in determining disease progression is outlined along with important cell-vessel interactions which combined together account for a great proportion of Hanahan and Weinberg's Hallmarks of Cancer.

一旦癌症开始,正常细胞突变为恶性细胞,实体肿瘤就会在其微环境中生长、发展和扩散,侵入局部组织;随着病情的发展,癌细胞在身体周围迁移,导致远处继发性肿瘤的形成。肿瘤与其微环境之间的相互作用驱动了这一系列事件,这些事件对人类宿主/患者具有毁灭性的后果,如果不是致命的后果。在这些相互作用中,生物力学相互作用是一个重要组成部分。在这篇综述文章中,通过数学和计算肿瘤学社区的建模工作,讨论了关键的生物力学关系。主要的焦点是直接的,自然地,以格子为基础的,基于力的实体肿瘤生长和发展模型。在这样的模型中,癌细胞对之间的相互作用(以及细胞和肿瘤微环境的其他结构之间的相互作用)是由力控制的。这些力是斥力和附着力,通常通过接触力学的扩展赫兹模型或使用约翰逊-肯德尔-罗伯茨理论来建模,这两种力在这里都进行了讨论。细胞外基质在决定疾病进展中的作用与重要的细胞-血管相互作用一起概述,这些相互作用结合在一起占Hanahan和Weinberg的癌症标志的很大比例。
{"title":"Biomechanical modelling of cancer: Agent-based force-based models of solid tumours within the context of the tumour microenvironment","authors":"Cicely K. Macnamara","doi":"10.1002/cso2.1018","DOIUrl":"10.1002/cso2.1018","url":null,"abstract":"<p>Once cancer is initiated, with normal cells mutated into malignant ones, a solid tumour grows, develops and spreads within its microenvironment invading the local tissue; the disease progresses and the cancer cells migrate around the body leading to metastasis, the formation of distant secondary tumours. Interactions between the tumour and its microenvironment drive this cascade of events which have devastating, if not fatal, consequences for the human host/patient. Among these interactions, biomechanical interactions are a vital component. In this review paper, key biomechanical relationships are discussed through a presentation of modelling efforts by the mathematical and computational oncology community. The main focus is directed, naturally, towards lattice-free agent-based, force-based models of solid tumour growth and development. In such models, interactions between pairs of cancer cells (as well as between cells and other structures of the tumour microenvironment) are governed by forces. These forces are ones of repulsion and adhesion, and are typically modelled via either an extended Hertz model of contact mechanics or using Johnson–Kendal–Roberts theory, both of which are discussed here. The role of the extracellular matrix in determining disease progression is outlined along with important cell-vessel interactions which combined together account for a great proportion of Hanahan and Weinberg's <i>Hallmarks of Cancer</i>.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-05-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1018","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42339163","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: 12
Boolean dynamic modeling of cancer signaling networks: Prognosis, progression, and therapeutics 癌症信号网络的布尔动态建模:预后、进展和治疗
Pub Date : 2021-05-01 DOI: 10.1002/cso2.1017
Shubhank Sherekar, Ganesh A. Viswanathan

Cancer is a multifactorial disease. Aberrant functioning of the underlying complex signaling network that orchestrates cellular response to external or internal cues governs incidence, progression, and recurrence of cancer. Detailed understanding of cancer's etiology can offer useful insights into arriving at novel therapeutic and disease management strategies. Such an understanding for most cancers is currently limited due to unavailability of a predictive large-scale, integrated signaling model accounting for all tumor orchestrating factors. We suggest that the potential of Boolean dynamic (BD) modeling approaches, though qualitative, can be harnessed for developing holistic models capturing multi-scale, multi-cellular signaling processes involved in cancer incidence and progression. We believe that constraining such an integrated BD model with variety of omics data at different scales from laboratory and clinical settings could offer deeper insights into causal mechanisms governing the disease leading to better prognosis. We review the recent literature employing different BD modeling strategies to model variety of cancer signaling programs leading to identification of cancer-specific prognostic markers such as SMAD proteins, which may also serve as early predictors of tumor cells hijacking the epithelial-mesenchymal plasticity program. In silico simulations of BD models of different cancer signaling networks combined with attractor landscape analysis and validated with experimental data predicted the nature of short- and long-term response of standard targeted therapeutic agents such as Nutlin-3, a small molecule inhibitor for p53-MDM2 interaction. BD simulations also offered a mechanistic view of emerging resistance to drugs such as Trastuzumab for HER+ breast cancer, analysis of which suggested new combination therapies to circumvent them. We believe future improvements in BD modeling techniques, and tools can lead to development of a comprehensive platform that can drive holistic approaches toward better decision-making in the clinical settings, and thereby help identify novel therapeutic strategies for improved cancer treatment at personalised levels.

癌症是一种多因素疾病。调控细胞对外部或内部信号反应的潜在复杂信号网络的异常功能控制着癌症的发生、进展和复发。详细了解癌症的病因可以提供有用的见解,以达到新的治疗和疾病管理策略。对于大多数癌症的这种理解目前是有限的,因为没有一个可预测的大规模、综合的信号模型来解释所有肿瘤协调因素。我们建议,布尔动态(BD)建模方法的潜力,虽然定性,可以用于开发整体模型,捕获涉及癌症发病率和进展的多尺度,多细胞信号传导过程。我们相信,结合来自实验室和临床环境的不同规模的各种组学数据来约束这种集成的双相障碍模型,可以更深入地了解控制疾病的因果机制,从而获得更好的预后。我们回顾了最近的文献,采用不同的BD建模策略来模拟各种癌症信号程序,从而确定癌症特异性预后标志物,如SMAD蛋白,它也可以作为肿瘤细胞劫持上皮-间质可塑性程序的早期预测因子。结合吸引子景观分析和实验数据验证,对不同癌症信号网络的BD模型进行了计算机模拟,预测了标准靶向治疗剂(如p53-MDM2相互作用的小分子抑制剂Nutlin-3)的短期和长期反应性质。BD模拟还提供了对HER+乳腺癌的曲妥珠单抗等药物出现耐药性的机制观点,分析表明可以采用新的联合疗法来规避它们。我们相信未来BD建模技术和工具的改进可以导致一个综合平台的发展,该平台可以推动整体方法在临床环境中做出更好的决策,从而帮助确定新的治疗策略,以改善个性化水平的癌症治疗。
{"title":"Boolean dynamic modeling of cancer signaling networks: Prognosis, progression, and therapeutics","authors":"Shubhank Sherekar,&nbsp;Ganesh A. Viswanathan","doi":"10.1002/cso2.1017","DOIUrl":"10.1002/cso2.1017","url":null,"abstract":"<p>Cancer is a multifactorial disease. Aberrant functioning of the underlying complex signaling network that orchestrates cellular response to external or internal cues governs incidence, progression, and recurrence of cancer. Detailed understanding of cancer's etiology can offer useful insights into arriving at novel therapeutic and disease management strategies. Such an understanding for most cancers is currently limited due to unavailability of a predictive large-scale, integrated signaling model accounting for all tumor orchestrating factors. We suggest that the potential of Boolean dynamic (BD) modeling approaches, though qualitative, can be harnessed for developing holistic models capturing multi-scale, multi-cellular signaling processes involved in cancer incidence and progression. We believe that constraining such an integrated BD model with variety of omics data at different scales from laboratory and clinical settings could offer deeper insights into causal mechanisms governing the disease leading to better prognosis. We review the recent literature employing different BD modeling strategies to model variety of cancer signaling programs leading to identification of cancer-specific prognostic markers such as SMAD proteins, which may also serve as early predictors of tumor cells hijacking the epithelial-mesenchymal plasticity program. <i>In silico</i> simulations of BD models of different cancer signaling networks combined with attractor landscape analysis and validated with experimental data predicted the nature of short- and long-term response of standard targeted therapeutic agents such as Nutlin-3, a small molecule inhibitor for p53-MDM2 interaction. BD simulations also offered a mechanistic view of emerging resistance to drugs such as Trastuzumab for HER+ breast cancer, analysis of which suggested new combination therapies to circumvent them. We believe future improvements in BD modeling techniques, and tools can lead to development of a comprehensive platform that can drive holistic approaches toward better decision-making in the clinical settings, and thereby help identify novel therapeutic strategies for improved cancer treatment at personalised levels.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1017","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43754769","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: 10
Computational immune infiltration analysis of pediatric high-grade gliomas (pHGGs) reveals differences in immunosuppression and prognosis by tumor location 小儿高级别胶质瘤(pHGGs)的计算免疫浸润分析揭示了不同肿瘤部位的免疫抑制和预后差异
Pub Date : 2021-03-08 DOI: 10.1002/cso2.1016
Cavan P. Bailey, Ruiping Wang, Mary Figueroa, Shaojun Zhang, Linghua Wang, Joya Chandra

Immunotherapy for cancer has moved from pre-clinical hypothesis to successful clinical application in the past 15 years. However, not all cancers have shown response rates in clinical trials for these new agents. igh-grade gliomas, in particular, have proved exceedingly refractory to immunotherapy. In adult patients, there has been much investigation into these failures, and researchers have concluded that an immunosuppressive microenvironment combined with low mutational burden renders adult glioblastomas “immune cold.” Pediatric cancer patients develop gliomas at a higher rate per malignancy than adults, and their brain tumors bear even fewer mutations. These tumors can also develop in more diverse locations in the brain, beyond the cerebral hemispheres seen in adults, including in the brainstem where critical motor functions are controlled. While adult brain tumor immune infiltration has been extensively profiled from surgical resections, this is not possible for brainstem tumors that can only be sampled at autopsy. Given these limitations, there is a dearth of information on immune cells and their therapeutic and prognostic impact in pediatric high-grade gliomas (pHGGs), including hemispheric tumors in addition to brainstem. In this report, we use computational methods to examine immune infiltrate in pHGGs and discover distinct immune patterns between hemispheric and brainstem tumors. In hemispheric tumors, we find positive prognostic associations for regulatory T-cells, memory B-cells, eosinophils, and dendritic cells, but not in brainstem tumors. These differences suggest that immunotherapeutic approaches must be cognizant of pHGG tumor location and tailored for optimum efficacy.

在过去的15年里,免疫治疗癌症已经从临床前的假设发展到成功的临床应用。然而,并不是所有的癌症在这些新药的临床试验中都显示出反应率。特别是高级别胶质瘤,已经证明对免疫治疗非常难治。在成人患者中,对这些失败进行了大量调查,研究人员得出结论,免疫抑制微环境与低突变负担相结合,使成人胶质母细胞瘤“免疫冷”。儿童癌症患者患胶质瘤的几率比成人高,而且他们的脑肿瘤发生的突变更少。这些肿瘤也可以在大脑的更多不同部位发展,除了在成人中看到的大脑半球,包括在控制关键运动功能的脑干。虽然成人脑肿瘤免疫浸润已被广泛地描述为手术切除,但这对于脑干肿瘤是不可能的,因为脑干肿瘤只能在尸检中取样。鉴于这些局限性,关于免疫细胞及其在儿童高级别胶质瘤(pHGGs)中的治疗和预后影响的信息缺乏,包括脑干以外的半球肿瘤。在本报告中,我们使用计算方法检查pHGGs中的免疫浸润,并发现半球和脑干肿瘤之间不同的免疫模式。在半球肿瘤中,我们发现调节性t细胞、记忆性b细胞、嗜酸性粒细胞和树突状细胞与预后呈正相关,但在脑干肿瘤中没有。这些差异表明,免疫治疗方法必须认识到pHGG肿瘤的位置和量身定制的最佳疗效。
{"title":"Computational immune infiltration analysis of pediatric high-grade gliomas (pHGGs) reveals differences in immunosuppression and prognosis by tumor location","authors":"Cavan P. Bailey,&nbsp;Ruiping Wang,&nbsp;Mary Figueroa,&nbsp;Shaojun Zhang,&nbsp;Linghua Wang,&nbsp;Joya Chandra","doi":"10.1002/cso2.1016","DOIUrl":"10.1002/cso2.1016","url":null,"abstract":"<p>Immunotherapy for cancer has moved from pre-clinical hypothesis to successful clinical application in the past 15 years. However, not all cancers have shown response rates in clinical trials for these new agents. igh-grade gliomas, in particular, have proved exceedingly refractory to immunotherapy. In adult patients, there has been much investigation into these failures, and researchers have concluded that an immunosuppressive microenvironment combined with low mutational burden renders adult glioblastomas “immune cold.” Pediatric cancer patients develop gliomas at a higher rate per malignancy than adults, and their brain tumors bear even fewer mutations. These tumors can also develop in more diverse locations in the brain, beyond the cerebral hemispheres seen in adults, including in the brainstem where critical motor functions are controlled. While adult brain tumor immune infiltration has been extensively profiled from surgical resections, this is not possible for brainstem tumors that can only be sampled at autopsy. Given these limitations, there is a dearth of information on immune cells and their therapeutic and prognostic impact in pediatric high-grade gliomas (pHGGs), including hemispheric tumors in addition to brainstem. In this report, we use computational methods to examine immune infiltrate in pHGGs and discover distinct immune patterns between hemispheric and brainstem tumors. In hemispheric tumors, we find positive prognostic associations for regulatory T-cells, memory B-cells, eosinophils, and dendritic cells, but not in brainstem tumors. These differences suggest that immunotherapeutic approaches must be cognizant of pHGG tumor location and tailored for optimum efficacy.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1016","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39581241","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: 4
Mechanistic insights into the heterogeneous response to anti-VEGF treatment in tumors 肿瘤抗vegf治疗的异质性反应机制
Pub Date : 2021-03-02 DOI: 10.1002/cso2.1013
Ding Li, Stacey D. Finley

Vascular endothelial growth factor (VEGF) is a strong promoter of angiogenesis in tumors, and anti-VEGF treatment, such as a humanized antibody to VEGF, is clinically used as a monotherapy or in combination with chemotherapy to treat cancer patients. However, this approach is not effective in all patients or cancer types. To better understand the heterogeneous responses to anti-VEGF and the synergy between anti-VEGF and other anticancer therapies, we constructed a computational model characterizing angiogenesis-mediated growth of in vivo mouse tumor xenografts. The model captures VEGF-mediated cross-talk between tumor cells and endothelial cells and is able to predict the details of molecular- and cellular-level dynamics. The model predictions of tumor growth in response to anti-VEGF closely match the quantitative measurements from multiple preclinical mouse studies. We applied the model to investigate the effects of VEGF-targeted treatment on tumor cells and endothelial cells. We identified that tumors with lower tumor cell growth rate and higher carrying capacity have a stronger response to anti-VEGF treatment. The predictions indicate that the variation of tumor cell growth rate can be a main reason for the experimentally observed heterogeneous response to anti-VEGF. In addition, our simulation results suggest a new synergy mechanism where anticancer therapy can enhance anti-VEGF simply through reducing the tumor cell growth rate. Overall, this work generates novel insights into the heterogeneous response to anti-VEGF treatment and the synergy of anti-VEGF with other therapies, providing a tool that be further used to test and optimize anticancer therapy.

血管内皮生长因子(Vascular endothelial growth factor, VEGF)是肿瘤血管生成的强有力的促进因子,抗VEGF治疗如针对VEGF的人源化抗体,在临床上作为单一疗法或联合化疗治疗癌症患者。然而,这种方法并不是对所有的病人或癌症类型都有效。为了更好地了解抗vegf的异质性反应以及抗vegf与其他抗癌疗法之间的协同作用,我们构建了一个表征血管生成介导的小鼠肿瘤异种移植物体内生长的计算模型。该模型捕获肿瘤细胞和内皮细胞之间vegf介导的串扰,并能够预测分子和细胞水平动力学的细节。该模型预测肿瘤生长对抗vegf的反应与多个临床前小鼠研究的定量测量结果密切匹配。我们应用该模型研究vegf靶向治疗对肿瘤细胞和内皮细胞的影响。我们发现肿瘤细胞生长速率低、携带能力高的肿瘤对抗vegf治疗的反应更强。这些预测表明,肿瘤细胞生长速率的变化可能是实验观察到的抗vegf异质性反应的主要原因。此外,我们的模拟结果提示了一种新的协同机制,即抗癌治疗可以通过降低肿瘤细胞的生长速度来增强抗vegf。总的来说,这项工作对抗vegf治疗的异质反应以及抗vegf与其他治疗的协同作用产生了新的见解,为进一步测试和优化抗癌治疗提供了一个工具。
{"title":"Mechanistic insights into the heterogeneous response to anti-VEGF treatment in tumors","authors":"Ding Li,&nbsp;Stacey D. Finley","doi":"10.1002/cso2.1013","DOIUrl":"10.1002/cso2.1013","url":null,"abstract":"<p>Vascular endothelial growth factor (VEGF) is a strong promoter of angiogenesis in tumors, and anti-VEGF treatment, such as a humanized antibody to VEGF, is clinically used as a monotherapy or in combination with chemotherapy to treat cancer patients. However, this approach is not effective in all patients or cancer types. To better understand the heterogeneous responses to anti-VEGF and the synergy between anti-VEGF and other anticancer therapies, we constructed a computational model characterizing angiogenesis-mediated growth of <i>in vivo</i> mouse tumor xenografts. The model captures VEGF-mediated cross-talk between tumor cells and endothelial cells and is able to predict the details of molecular- and cellular-level dynamics. The model predictions of tumor growth in response to anti-VEGF closely match the quantitative measurements from multiple preclinical mouse studies. We applied the model to investigate the effects of VEGF-targeted treatment on tumor cells and endothelial cells. We identified that tumors with lower tumor cell growth rate and higher carrying capacity have a stronger response to anti-VEGF treatment. The predictions indicate that the variation of tumor cell growth rate can be a main reason for the experimentally observed heterogeneous response to anti-VEGF. In addition, our simulation results suggest a new synergy mechanism where anticancer therapy can enhance anti-VEGF simply through reducing the tumor cell growth rate. Overall, this work generates novel insights into the heterogeneous response to anti-VEGF treatment and the synergy of anti-VEGF with other therapies, providing a tool that be further used to test and optimize anticancer therapy.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1013","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49477916","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: 4
Investigating epithelial-mesenchymal heterogeneity of tumors and circulating tumor cells with transcriptomic analysis and biophysical modeling 利用转录组学分析和生物物理模型研究肿瘤和循环肿瘤细胞的上皮-间质异质性
Pub Date : 2021-03-01 DOI: 10.1002/cso2.1015
Federico Bocci, Susmita Mandal, Tanishq Tejaswi, Mohit Kumar Jolly

Cellular heterogeneity along the epithelial-mesenchymal plasticity (EMP) spectrum is a paramount feature observed in tumors and circulating tumor cells (CTCs). High-throughput techniques now offer unprecedented details on this variability at a single-cell resolution. Yet, there is no current consensus about how EMP in tumors propagates to that in CTCs. To investigate the relationship between EMP-associated heterogeneity of tumors and that of CTCs, we integrated transcriptomic analysis and biophysical modeling. We apply three epithelial-mesenchymal transition (EMT) scoring metrics to multiple tumor samples and CTC datasets from several cancer types. Moreover, we develop a biophysical model that couples EMT-associated phenotypic switching in a primary tumor with cell migration. Finally, we integrate EMT transcriptomic analysis and in silico modeling to evaluate the predictive power of several measurements of tumor aggressiveness, including tumor EMT score, CTC EMT score, fraction of CTC clusters found in circulation, and CTC cluster size distribution. Analysis of high-throughput datasets reveals a pronounced heterogeneity without a well-defined relation between EMT traits in tumors and CTCs. Moreover, mathematical modeling predicts different phases where CTCs can be less, equally, or more mesenchymal than primary tumor depending on the dynamics of phenotypic transition and cell migration. Consistently, various datasets of CTC cluster size distribution from different cancer types are fitted onto different regimes of the model. By further constraining the model with experimental measurements of tumor EMT score, CTC EMT score, and fraction of CTC cluster in bloodstream, we show that none of these assays alone can provide sufficient information to predict the other variables. In conclusion, we propose that the relationship between EMT progression in tumors and CTCs can be variable, and in general, predicting one from the other may not be as straightforward as tacitly assumed.

上皮-间充质可塑性(EMP)谱上的细胞异质性是肿瘤和循环肿瘤细胞(ctc)的一个重要特征。高通量技术现在以单细胞分辨率提供了这种可变性的前所未有的细节。然而,目前对于肿瘤中的EMP如何传播到ctc中尚无共识。为了研究emp相关肿瘤异质性与ctc异质性之间的关系,我们整合了转录组学分析和生物物理模型。我们将三个上皮-间质转化(EMT)评分指标应用于多个肿瘤样本和来自几种癌症类型的CTC数据集。此外,我们开发了一种生物物理模型,将原发性肿瘤中emt相关的表型转换与细胞迁移耦合在一起。最后,我们整合了EMT转录组学分析和计算机建模,以评估肿瘤侵袭性的几种测量方法的预测能力,包括肿瘤EMT评分、CTC EMT评分、循环中发现的CTC簇的比例和CTC簇大小分布。对高通量数据集的分析显示,肿瘤中EMT特征与ctc之间存在明显的异质性,但没有明确的关系。此外,根据表型转变和细胞迁移的动态,数学模型预测了不同的阶段,ctc可能比原发肿瘤更少、相同或更多的间质性。一致地,来自不同癌症类型的CTC簇大小分布的各种数据集被拟合到模型的不同制度上。通过进一步用肿瘤EMT评分、CTC EMT评分和血液中CTC簇的分数的实验测量来约束模型,我们发现这些分析都不能单独提供足够的信息来预测其他变量。总之,我们认为肿瘤中EMT进展与ctc之间的关系可能是可变的,一般来说,预测一个与另一个之间的关系可能不像默认的那样简单。
{"title":"Investigating epithelial-mesenchymal heterogeneity of tumors and circulating tumor cells with transcriptomic analysis and biophysical modeling","authors":"Federico Bocci,&nbsp;Susmita Mandal,&nbsp;Tanishq Tejaswi,&nbsp;Mohit Kumar Jolly","doi":"10.1002/cso2.1015","DOIUrl":"https://doi.org/10.1002/cso2.1015","url":null,"abstract":"<p>Cellular heterogeneity along the epithelial-mesenchymal plasticity (EMP) spectrum is a paramount feature observed in tumors and circulating tumor cells (CTCs). High-throughput techniques now offer unprecedented details on this variability at a single-cell resolution. Yet, there is no current consensus about how EMP in tumors propagates to that in CTCs. To investigate the relationship between EMP-associated heterogeneity of tumors and that of CTCs, we integrated transcriptomic analysis and biophysical modeling. We apply three epithelial-mesenchymal transition (EMT) scoring metrics to multiple tumor samples and CTC datasets from several cancer types. Moreover, we develop a biophysical model that couples EMT-associated phenotypic switching in a primary tumor with cell migration. Finally, we integrate EMT transcriptomic analysis and in silico modeling to evaluate the predictive power of several measurements of tumor aggressiveness, including tumor EMT score, CTC EMT score, fraction of CTC clusters found in circulation, and CTC cluster size distribution. Analysis of high-throughput datasets reveals a pronounced heterogeneity without a well-defined relation between EMT traits in tumors and CTCs. Moreover, mathematical modeling predicts different phases where CTCs can be less, equally, or more mesenchymal than primary tumor depending on the dynamics of phenotypic transition and cell migration. Consistently, various datasets of CTC cluster size distribution from different cancer types are fitted onto different regimes of the model. By further constraining the model with experimental measurements of tumor EMT score, CTC EMT score, and fraction of CTC cluster in bloodstream, we show that none of these assays alone can provide sufficient information to predict the other variables. In conclusion, we propose that the relationship between EMT progression in tumors and CTCs can be variable, and in general, predicting one from the other may not be as straightforward as tacitly assumed.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1015","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"137460925","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
Relating prostate-specific antigen leakage with vascular tumor growth in a mathematical model of prostate cancer response to androgen deprivation 前列腺癌对雄激素剥夺反应的数学模型中前列腺特异性抗原泄漏与血管肿瘤生长的关系
Pub Date : 2021-03-01 DOI: 10.1002/cso2.1014
Johnna P. Barnaby, Inmaculada C. Sorribes, Harsh Vardhan Jain

The use of prostate-specific antigen (PSA) as a prognostic indicator for prostate cancer (PCa) patients is controversial, especially since it has been shown to correlate poorly with tumor burden. The poor quality of PSA as a biomarker could be explained by current guidelines not accounting for the mechanism by which it enters circulation. Given that mature blood vessels are relatively impermeable to it, we hypothesize that immature and leaky blood vessels, formed under angiogenic cues in a hypoxic tumor, facilitate PSA extravasation into circulation. To explore our hypothesis, we develop a nonlinear dynamical systems model describing the vascular growth of PCa, that explicitly links PSA leakage into circulation with changes in intra-tumoral oxygen tension and vessel permeability. The model is calibrated versus serum PSA and tumor burden time-courses from a mouse xenograft model of castration resistant PCa response to androgen deprivation. The model recapitulates the experimentally observed and – counterintuitive – phenomenon of increasing tumor burden despite decreasing serum PSA levels. The validated model is then extended to the human scale by incorporating patient-specific parameters and fitting individual PSA time-courses from patients with biochemically failing PCa. Our results highlight the limitations of using time to PSA failure as a clinical indicator of androgen deprivation efficacy. We propose an alternative indicator, namely a treatment efficacy index, for patients with castration resistant disease, to identify who would benefit most from enhanced androgen deprivation. A critical challenge in PCa therapeutics is quantifying the relationship between serum PSA and tumor burden. Our results underscore the potential of mathematical modeling in understanding the limitations of serum PSA as a prognostic indicator. Finally, we provide a means of augmenting PSA time-courses in the diagnostic process, with changes in intra-tumoral vascularity and vascular architecture.

使用前列腺特异性抗原(PSA)作为前列腺癌(PCa)患者的预后指标是有争议的,特别是因为它已被证明与肿瘤负荷相关性很差。PSA作为生物标志物的低质量可以解释为目前的指南没有考虑其进入循环的机制。鉴于成熟血管对PSA的渗透性相对较差,我们假设在缺氧肿瘤血管生成提示下形成的未成熟和渗漏血管促进PSA外渗进入循环。为了探索我们的假设,我们建立了一个描述前列腺癌血管生长的非线性动态系统模型,明确地将PSA渗漏到循环中与肿瘤内氧张力和血管渗透性的变化联系起来。该模型是根据抗去势PCa对雄激素剥夺反应的小鼠异种移植模型的血清PSA和肿瘤负荷时间过程校准的。该模型概括了实验观察到的与直觉相反的现象,即尽管血清PSA水平降低,但肿瘤负荷仍在增加。然后,通过纳入患者特异性参数和拟合生物化学失败的PCa患者的个体PSA时间过程,将验证模型扩展到人体尺度。我们的研究结果强调了将PSA失败的时间作为雄激素剥夺疗效的临床指标的局限性。我们提出一个替代指标,即治疗效果指数,去势抵抗疾病的患者,以确定谁将从加强雄激素剥夺中获益最多。前列腺癌治疗的一个关键挑战是量化血清PSA与肿瘤负荷之间的关系。我们的结果强调了数学模型在理解血清PSA作为预后指标的局限性方面的潜力。最后,我们提供了一种在诊断过程中增加PSA时间过程的方法,随着肿瘤内血管和血管结构的变化。
{"title":"Relating prostate-specific antigen leakage with vascular tumor growth in a mathematical model of prostate cancer response to androgen deprivation","authors":"Johnna P. Barnaby,&nbsp;Inmaculada C. Sorribes,&nbsp;Harsh Vardhan Jain","doi":"10.1002/cso2.1014","DOIUrl":"10.1002/cso2.1014","url":null,"abstract":"<p>The use of prostate-specific antigen (PSA) as a prognostic indicator for prostate cancer (PCa) patients is controversial, especially since it has been shown to correlate poorly with tumor burden. The poor quality of PSA as a biomarker could be explained by current guidelines not accounting for the mechanism by which it enters circulation. Given that mature blood vessels are relatively impermeable to it, we hypothesize that immature and leaky blood vessels, formed under angiogenic cues in a hypoxic tumor, facilitate PSA extravasation into circulation. To explore our hypothesis, we develop a nonlinear dynamical systems model describing the vascular growth of PCa, that explicitly links PSA leakage into circulation with changes in intra-tumoral oxygen tension and vessel permeability. The model is calibrated versus serum PSA and tumor burden time-courses from a mouse xenograft model of castration resistant PCa response to androgen deprivation. The model recapitulates the experimentally observed and – counterintuitive – phenomenon of increasing tumor burden despite decreasing serum PSA levels. The validated model is then extended to the human scale by incorporating patient-specific parameters and fitting individual PSA time-courses from patients with biochemically failing PCa. Our results highlight the limitations of using time to PSA failure as a clinical indicator of androgen deprivation efficacy. We propose an alternative indicator, namely a treatment efficacy index, for patients with castration resistant disease, to identify who would benefit most from enhanced androgen deprivation. A critical challenge in PCa therapeutics is quantifying the relationship between serum PSA and tumor burden. Our results underscore the potential of mathematical modeling in understanding the limitations of serum PSA as a prognostic indicator. Finally, we provide a means of augmenting PSA time-courses in the diagnostic process, with changes in intra-tumoral vascularity and vascular architecture.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1014","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41629096","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: 6
Are all models wrong? 是不是所有的模型都错了?
Pub Date : 2021-01-15 DOI: 10.1002/cso2.1008
Heiko Enderling, Olaf Wolkenhauer

Mathematical modeling in cancer is enjoying a rapid expansion [1]. For collegial discussion across disciplines, many—if not all of us—have used the aphorism that “All models are wrong, but some are useful” [2]. This has been a convenient approach to justify and communicate the praxis of modeling. This is to suggest that the usefulness of a model is not measured by the accuracy of representation but how well it supports the generation, testing, and refinement of hypotheses. A key insight is not to focus on the model as an outcome, but to consider the modeling process and simulated model predictions as “ways of thinking” about complex nonlinear dynamical systems [3]. Here, we discuss the convoluted interpretation of models being wrong in the arena of predictive modeling.

All models are wrong, but some are useful” emphasizes the value of abstraction in order to gain insight. While abstraction clearly implies misrepresentation, it allows to explicitly define model assumptions and interpret model results within these limitations – Truth emerges more readily from error than from confusion [4]. It is thus the process of modeling and the discussions about model assumptions that are often considered most valuable in interdisciplinary research. They provide a way of thinking about complex systems and mechanisms underlying observations. Abstractions are being made in cancer biology for every experiment in each laboratory around the world. In vitro cell lines or in vivo mouse experiments are abstractions of complex adaptive evolving human cancers in the complex adaptive dynamic environment called the patient. These "wet lab" experiments akin to "dry lab" mathematical models offer confirmation or refutation of hypotheses and results, which have to be prospectively evaluated in clinical trials before conclusions can be generalized beyond the abstracted assumptions. The key for any model—mathematical, biological, or clinical—to succeed is an iterative cycle of data-driven modeling and model-driven experimentation [5, 6]. The value of such an effort lies in the insights about mechanisms that can then be attributed to the considered variables [7]. With simplified representations of a system one can learn about the emergence of general patterns, like the occurrence of oscillations, bistability, or chaos [8-10].

In this context, Alan Turing framed the purpose of a mathematical model in his seminal paper about “The chemical basis of morphogenesis” [11] with “This model will be a simplification and an idealization, and consequently a falsification. It is to be hoped that the features retained for discussion are those of greatest importance in the present state of knowledge.” For many mathematical biology models that are built to explore, test, and generate hypotheses about emerging dynamics, this remains tru

癌症领域的数学建模正在迅速发展[1]。对于跨学科的合作讨论,许多人——如果不是我们所有人——都使用了“所有模型都是错误的,但有些模型是有用的”这句格言[2]。这是证明和交流建模实践的一种方便的方法。这表明,模型的有用性不是通过表示的准确性来衡量的,而是通过它对假设的生成、测试和改进的支持程度来衡量的。一个关键的见解是,不要将模型作为结果来关注,而是将建模过程和模拟模型预测视为复杂非线性动力系统的“思维方式”[3]。在这里,我们将讨论预测建模领域中对模型错误的复杂解释。“所有的模型都是错误的,但有些是有用的”强调了为了获得洞察力而抽象的价值。虽然抽象显然意味着错误表述,但它允许明确定义模型假设并在这些限制内解释模型结果——真理更容易从错误中出现,而不是从混乱中出现[4]。因此,在跨学科研究中,建模过程和关于模型假设的讨论通常被认为是最有价值的。它们提供了一种思考复杂系统和潜在观察机制的方法。世界上每个实验室的每个实验都在对癌症生物学进行抽象。体外细胞系或体内小鼠实验是人类癌症在称为患者的复杂适应动态环境中复杂适应进化的抽象。这些类似于“干实验室”数学模型的“湿实验室”实验提供了对假设和结果的证实或反驳,这些假设和结果必须在临床试验中进行前瞻性评估,然后才能在抽象假设之外推广结论。任何模型——数学、生物或临床——成功的关键是数据驱动的建模和模型驱动的实验的迭代循环[5,6]。这种努力的价值在于对机制的洞察,然后可以归因于所考虑的变量[7]。通过系统的简化表示,人们可以了解一般模式的出现,如振荡、双稳态或混沌的发生[8-10]。在这种背景下,艾伦·图灵在他的开创性论文《形态发生的化学基础》[11]中提出了数学模型的目的,“这个模型将是一个简化和理想化,因此是一个证伪。”我们希望保留下来讨论的特征是在目前的知识状态下最重要的特征"对于许多用于探索、测试和生成关于新兴动力学的假设的数学生物学模型来说,这仍然是正确的。“错误的模型”允许我们重新评估我们的假设,从这些讨论中吸取的教训可以帮助制定修订的模型,并提高我们对潜在动力学的理解。然而,数学肿瘤学模型不仅用于模拟复杂系统的紧急特性,以生成、测试和完善假设,而且越来越多地用于预测——通常是单个癌症患者对特定治疗的反应[1]。对于预测建模来说,“所有的模型都是错的”这句格言显得有些尴尬。在预测建模领域,有用的模型不应该是错误的。预测建模应用的一个主要障碍,一般来说,特别是在肿瘤学领域,是模型目的和预测不确定性的沟通,以及最终用户如何解释可能性和风险。由于复杂的自适应进化系统的可用数据有限,当数据中未表示的事件主导后续行为(例如未在预处理动态中表示的治疗耐药性的出现)时,“预测失败”很常见。如果预测模型是在历史数据上训练的,但在多个时间点上很少有患者特定的数据,那么预测模型在肿瘤学中可以发挥什么作用?以有限的数据为基础的数学模型的计算机模拟仅仅是将疾病的发展轨迹可视化。然后,可以使用多个合理的参数组合,从单个模型或具有相互竞争的假设和可能重要因素的不同权重的多个模型,分析可能的轨迹,从而做出预测。虽然在某些领域,如飓风轨迹预测,我们相信数学模型并接受其固有的、有充分记录的预测不确定性[12],但在涉及个人健康时,必须改善模型能做什么和不能做什么的沟通。“没有什么比预测未来更困难的了”,虽然与预测相关的不确定性上升得很快,但我们仍然可以在这个模型中找到用处。
{"title":"Are all models wrong?","authors":"Heiko Enderling,&nbsp;Olaf Wolkenhauer","doi":"10.1002/cso2.1008","DOIUrl":"10.1002/cso2.1008","url":null,"abstract":"<p>Mathematical modeling in cancer is enjoying a rapid expansion [<span>1</span>]. For collegial discussion across disciplines, many—if not all of us—have used the aphorism that “<i>All models are wrong, but some are useful</i>” [<span>2</span>]. This has been a convenient approach to justify and communicate the praxis of modeling. This is to suggest that the <i>usefulness</i> of a model is not measured by the accuracy of representation but how well it supports the generation, testing, and refinement of hypotheses. A key insight is not to focus on the model as an outcome, but to consider the modeling process and simulated model predictions as “ways of thinking” about complex nonlinear dynamical systems [<span>3</span>]. Here, we discuss the convoluted interpretation of <i>models being wrong</i> in the arena of predictive modeling.</p><p>“<i>All models are wrong, but some are useful</i>” emphasizes the value of abstraction in order to gain insight. While abstraction clearly implies misrepresentation, it allows to explicitly define model assumptions and interpret model results within these limitations – <i>Truth emerges more readily from error than from confusion</i> [<span>4</span>]. It is thus the process of modeling and the discussions about model assumptions that are often considered most valuable in interdisciplinary research. They provide a way of thinking about complex systems and mechanisms underlying observations. Abstractions are being made in cancer biology for every experiment in each laboratory around the world. In vitro cell lines or in vivo mouse experiments are abstractions of complex adaptive evolving human cancers in the complex adaptive dynamic environment called the patient. These \"wet lab\" experiments akin to \"dry lab\" mathematical models offer confirmation or refutation of hypotheses and results, which have to be prospectively evaluated in clinical trials before conclusions can be generalized beyond the abstracted assumptions. The key for any model—mathematical, biological, or clinical—to succeed is an iterative cycle of data-driven modeling and model-driven experimentation [<span>5, 6</span>]. The value of such an effort lies in the insights about mechanisms that can then be attributed to the considered variables [<span>7</span>]. With simplified representations of a system one can learn about the emergence of general patterns, like the occurrence of oscillations, bistability, or chaos [<span>8-10</span>].</p><p>In this context, Alan Turing framed the purpose of a mathematical model in his seminal paper about “The chemical basis of morphogenesis” [<span>11</span>] with “<i>This model will be a simplification and an idealization, and consequently a falsification. It is to be hoped that the features retained for discussion are those of greatest importance in the present state of knowledge</i>.” For many mathematical biology models that are built to explore, test, and generate hypotheses about emerging dynamics, this remains tru","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1008","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"25372405","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: 48
Water as a reactant in the differential expression of proteins in cancer 水作为反应物在癌症中蛋白质的差异表达
Pub Date : 2021-01-12 DOI: 10.1002/cso2.1007
Jeffrey M. Dick

Introduction. How proteomes differ between normal tissue and tumor microenvironments is an important question for cancer biochemistry. Methods. More than 250 datasets for differentially expressed (up- and downregulated) proteins compiled from the literature were analyzed to calculate the stoichiometric hydration state, which represents the number of water molecules in theoretical mass-balance reactions to form the proteins from a set of basis species. Results. The analysis shows increased stoichiometric hydration state of differentially expressed proteins in cancer compared to normal tissue. In contrast, experiments with different cell types grown in 3D compared to monolayer culture, or exposed to hyperosmotic conditions under high salt or high glucose, cause proteomes to “dry out” as measured by decreased stoichiometric hydration state of the differentially expressed proteins. Conclusion. These findings reveal a basic physicochemical link between proteome composition and water content, which is elevated in many tumors and proliferating cells.

介绍。蛋白质组学在正常组织和肿瘤微环境之间的差异是癌症生物化学的一个重要问题。方法。从文献中编译了250多个差异表达(上调和下调)蛋白质的数据集,分析了这些数据集,以计算化学计量水合状态,水合状态代表了从一组基本物种形成蛋白质的理论质量平衡反应中水分子的数量。结果。分析表明,与正常组织相比,癌症中差异表达蛋白的化学计量水合状态增加。相反,与单层培养相比,在3D中培养不同类型的细胞,或暴露于高盐或高葡萄糖的高渗透条件下,通过降低差异表达蛋白的化学计量水合状态来测量,导致蛋白质组“干化”。结论。这些发现揭示了蛋白质组组成和水含量之间的基本物理化学联系,水含量在许多肿瘤和增殖细胞中升高。
{"title":"Water as a reactant in the differential expression of proteins in cancer","authors":"Jeffrey M. Dick","doi":"10.1002/cso2.1007","DOIUrl":"https://doi.org/10.1002/cso2.1007","url":null,"abstract":"<p><i>Introduction</i>. How proteomes differ between normal tissue and tumor microenvironments is an important question for cancer biochemistry. <i>Methods</i>. More than 250 datasets for differentially expressed (up- and downregulated) proteins compiled from the literature were analyzed to calculate the stoichiometric hydration state, which represents the number of water molecules in theoretical mass-balance reactions to form the proteins from a set of basis species. <i>Results</i>. The analysis shows increased stoichiometric hydration state of differentially expressed proteins in cancer compared to normal tissue. In contrast, experiments with different cell types grown in 3D compared to monolayer culture, or exposed to hyperosmotic conditions under high salt or high glucose, cause proteomes to “dry out” as measured by decreased stoichiometric hydration state of the differentially expressed proteins. <i>Conclusion</i>. These findings reveal a basic physicochemical link between proteome composition and water content, which is elevated in many tumors and proliferating cells.</p>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1007","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"137827523","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
Bayesian competing risk analysis: An application to nasopharyngeal carcinoma patients data 贝叶斯竞争风险分析在鼻咽癌患者资料中的应用
Pub Date : 2021-01-03 DOI: 10.1002/cso2.1006
Rakesh Kumar Saroj, K. Narasimha Murthy, Mukesh Kumar, Atanu Bhattacharjee, Kamalesh Kumar Patel

Background

The Cox proportional hazard (CPH) model is normally used to study the death event data. The presence of competing risk (CR) is often encountered in health data, hence it becomes difficult to manage time to event data in clinical study. Bayesian approach is considered to manage the CR events in clinical data.

Objectives

The objective of study is to find the predictors associated with overall survival of nasopharyngeal carcinoma (NPC) patients. Further, our purpose is to use a Bayesian model that can analyze time to event data in the presence of CR.

Methods

Total 245 patients with NPC were taken (https://www.ncbi.nlm.nih.gov/geo/). The sociodemographic and clinical variables were considered for analysis purposes. R software and openBUGS were used to overcome the computational problems of CPH and Bayesian models. The Markov chain Monte Carlo (MCMC) method was used to compute the regression coefficients of Bayesian model.

Results

The study shows that among NPC patients, the covariates chemotherapy, smoking, N-stage, and tumor site are associated with the higher risk for the deaths occurring in the cancer patients. The posterior mean estimates of proposed Bayesian model for significant factors have been obtained. The posterior mean and standard deviation estimates help to improve the survival of patients in the presence of CR.

Conclusions

It is very difficult to use the CR model with Bayesian approach in health research for nonstatistical researcher due to lack of information. This paper is dedicated to the application of Bayesian approach for CR analysis on NPC data.

背景Cox比例风险(CPH)模型通常用于研究死亡事件数据。健康数据中经常出现竞争风险(CR),因此临床研究中对事件时间数据的管理变得困难。贝叶斯方法被认为是处理临床数据中CR事件的方法。目的探讨鼻咽癌(NPC)患者总生存期的相关预测因素。此外,我们的目的是使用贝叶斯模型来分析CR存在时的事件时间数据。方法共收集245例NPC患者(https://www.ncbi.nlm.nih.gov/geo/)。为了分析目的,考虑了社会人口学和临床变量。利用R软件和openBUGS克服了CPH和贝叶斯模型的计算问题。采用马尔可夫链蒙特卡罗(MCMC)方法计算贝叶斯模型的回归系数。结果在鼻咽癌患者中,化疗、吸烟、n分期和肿瘤部位与肿瘤患者死亡风险增高相关。得到了贝叶斯模型对显著因子的后验均值估计。后验均值和标准差估计有助于提高CR存在时患者的生存率。结论由于信息的缺乏,非统计研究人员很难将贝叶斯方法的CR模型应用于健康研究。本文主要研究贝叶斯方法在NPC数据CR分析中的应用。
{"title":"Bayesian competing risk analysis: An application to nasopharyngeal carcinoma patients data","authors":"Rakesh Kumar Saroj,&nbsp;K. Narasimha Murthy,&nbsp;Mukesh Kumar,&nbsp;Atanu Bhattacharjee,&nbsp;Kamalesh Kumar Patel","doi":"10.1002/cso2.1006","DOIUrl":"10.1002/cso2.1006","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <h3> Background</h3>\u0000 \u0000 <p>The Cox proportional hazard (CPH) model is normally used to study the death event data. The presence of competing risk (CR) is often encountered in health data, hence it becomes difficult to manage time to event data in clinical study. Bayesian approach is considered to manage the CR events in clinical data.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Objectives</h3>\u0000 \u0000 <p>The objective of study is to find the predictors associated with overall survival of nasopharyngeal carcinoma (NPC) patients. Further, our purpose is to use a Bayesian model that can analyze time to event data in the presence of CR.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Methods</h3>\u0000 \u0000 <p>Total 245 patients with NPC were taken (https://www.ncbi.nlm.nih.gov/geo/). The sociodemographic and clinical variables were considered for analysis purposes. R software and openBUGS were used to overcome the computational problems of CPH and Bayesian models. The Markov chain Monte Carlo (MCMC) method was used to compute the regression coefficients of Bayesian model.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Results</h3>\u0000 \u0000 <p>The study shows that among NPC patients, the covariates chemotherapy, smoking, N-stage, and tumor site are associated with the higher risk for the deaths occurring in the cancer patients. The posterior mean estimates of proposed Bayesian model for significant factors have been obtained. The posterior mean and standard deviation estimates help to improve the survival of patients in the presence of CR.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Conclusions</h3>\u0000 \u0000 <p>It is very difficult to use the CR model with Bayesian approach in health research for nonstatistical researcher due to lack of information. This paper is dedicated to the application of Bayesian approach for CR analysis on NPC data.</p>\u0000 </section>\u0000 </div>","PeriodicalId":72658,"journal":{"name":"Computational and systems oncology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1002/cso2.1006","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"111288474","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
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