1
|
Li H, Huang Z, Guo C, Wang Y, Li B, Wang S, Bai N, Chen H, Xue J, Wang D, Zheng Z, Bing Z, Song Y, Xu Y, Huang G, Yu X, Li R, Fung KL, Li J, Song L, Zhu Z, Liu S, Liang N, Li S. Super multiple primary lung cancers harbor high-frequency BRAF and low-frequency EGFR mutations in the MAPK pathway. NPJ Precis Oncol 2024; 8:229. [PMID: 39384982 PMCID: PMC11464572 DOI: 10.1038/s41698-024-00726-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
The incidence of multiple primary lung cancer (MPLC) is increasing, with some of our surgical patients exhibiting numerous lesions. We defined lung cancer with five or more primary lesions as super MPLCs. Elucidating the genomic characteristics of this special MPLC subtype can help reduce disease burden and understand tumor evolution. In our cohort of synchronous super early-stage MPLCs (PUMCH-ssesMPLC), whole-exome sequencing on 130 resected malignant specimens from 18 patients provided comprehensive super-MPLC genomic landscapes. Mutations are enriched in PI3k-Akt and MAPK pathways. Their BRAF mutation frequency (31.5%) is significantly higher than MPLC with fewer lesions and early-stage single-lesion cancer, while EGFR mutations are significantly fewer (13.8%). As lesion counts increase, BRAF mutations gradually become dominant. Also, invasive lesions more tend to have classic super-MPLC mutation patterns. High-frequency BRAF mutations, especially Class II, and low-frequency EGFR mutations could be a reason for the limited effectiveness of targeted therapy in super-MPLC patients.
Collapse
Affiliation(s)
- Haochen Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Zhicheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Bowen Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Sha Wang
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, 210032, China
| | - Na Bai
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, 210032, China
| | - Hanlin Chen
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, 210032, China
| | - Jianchao Xue
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Daoyun Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhibo Zheng
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of International Medical Services, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yang Song
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuan Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guanghua Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiaoqing Yu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ruirui Li
- Department of Thoracic Surgery, Aviation General Hospital, Beijing, 100025, China
| | | | - Ji Li
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lan Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziwei Zhu
- Zhenyuan (Tianjin) Medical Technology Co. Ltd., Tianjin, 300385, China
| | - Songtao Liu
- Zhenyuan (Tianjin) Medical Technology Co. Ltd., Tianjin, 300385, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
2
|
Ponce-de-Leon M, Montagud A, Noël V, Meert A, Pradas G, Barillot E, Calzone L, Valencia A. PhysiBoSS 2.0: a sustainable integration of stochastic Boolean and agent-based modelling frameworks. NPJ Syst Biol Appl 2023; 9:54. [PMID: 37903760 PMCID: PMC10616087 DOI: 10.1038/s41540-023-00314-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/11/2023] [Indexed: 11/01/2023] Open
Abstract
In systems biology, mathematical models and simulations play a crucial role in understanding complex biological systems. Different modelling frameworks are employed depending on the nature and scales of the system under study. For instance, signalling and regulatory networks can be simulated using Boolean modelling, whereas multicellular systems can be studied using agent-based modelling. Herein, we present PhysiBoSS 2.0, a hybrid agent-based modelling framework that allows simulating signalling and regulatory networks within individual cell agents. PhysiBoSS 2.0 is a redesign and reimplementation of PhysiBoSS 1.0 and was conceived as an add-on that expands the PhysiCell functionalities by enabling the simulation of intracellular cell signalling using MaBoSS while keeping a decoupled, maintainable and model-agnostic design. PhysiBoSS 2.0 also expands the set of functionalities offered to the users, including custom models and cell specifications, mechanistic submodels of substrate internalisation and detailed control over simulation parameters. Together with PhysiBoSS 2.0, we introduce PCTK, a Python package developed for handling and processing simulation outputs, and generating summary plots and 3D renders. PhysiBoSS 2.0 allows studying the interplay between the microenvironment, the signalling pathways that control cellular processes and population dynamics, suitable for modelling cancer. We show different approaches for integrating Boolean networks into multi-scale simulations using strategies to study the drug effects and synergies in models of cancer cell lines and validate them using experimental data. PhysiBoSS 2.0 is open-source and publicly available on GitHub with several repositories of accompanying interoperable tools.
Collapse
Affiliation(s)
- Miguel Ponce-de-Leon
- Life Science, Barcelona Supercomputing Center (BSC), 1-3 Plaça Eusebi Güell, 08034, Barcelona, Spain
| | - Arnau Montagud
- Life Science, Barcelona Supercomputing Center (BSC), 1-3 Plaça Eusebi Güell, 08034, Barcelona, Spain
| | - Vincent Noël
- Institut Curie, Université PSL, 26 rue d'Ulm, 75248, Paris, France
- INSERM U900, Paris, France
- Mines ParisTech, Université PSL, Paris, France
| | - Annika Meert
- Life Science, Barcelona Supercomputing Center (BSC), 1-3 Plaça Eusebi Güell, 08034, Barcelona, Spain
| | - Gerard Pradas
- Life Science, Barcelona Supercomputing Center (BSC), 1-3 Plaça Eusebi Güell, 08034, Barcelona, Spain
| | - Emmanuel Barillot
- Institut Curie, Université PSL, 26 rue d'Ulm, 75248, Paris, France
- INSERM U900, Paris, France
- Mines ParisTech, Université PSL, Paris, France
| | - Laurence Calzone
- Institut Curie, Université PSL, 26 rue d'Ulm, 75248, Paris, France
- INSERM U900, Paris, France
- Mines ParisTech, Université PSL, Paris, France
| | - Alfonso Valencia
- Life Science, Barcelona Supercomputing Center (BSC), 1-3 Plaça Eusebi Güell, 08034, Barcelona, Spain.
- ICREA, 23 Passeig Lluís Companys, 08010, Barcelona, Spain.
| |
Collapse
|
3
|
Konrath F, Willenbrock M, Busse D, Scheidereit C, Wolf J. A computational model of the DNA damage-induced IKK/ NF-κB pathway reveals a critical dependence on irradiation dose and PARP-1. iScience 2023; 26:107917. [PMID: 37817938 PMCID: PMC10561052 DOI: 10.1016/j.isci.2023.107917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/01/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
The activation of IKK/NF-κB by genotoxic stress is a crucial process in the DNA damage response. Due to the anti-apoptotic impact of NF-κB, it can affect cell-fate decisions upon DNA damage and therefore interfere with tumor therapy-induced cell death. Here, we developed a dynamical model describing IKK/NF-κB signaling that faithfully reproduces quantitative time course data and enables a detailed analysis of pathway regulation. The approach elucidates a pathway topology with two hubs, where the first integrates signals from two DNA damage sensors and the second forms a coherent feedforward loop. The analyses reveal a critical role of the sensor protein PARP-1 in the pathway regulation. Introducing a method for calculating the impact of changes in individual components on pathway activity in a time-resolved manner, we show how irradiation dose influences pathway activation. Our results give a mechanistic understanding relevant for the interpretation of experimental and clinical studies.
Collapse
Affiliation(s)
- Fabian Konrath
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Michael Willenbrock
- Laboratory for Signal Transduction in Tumor Cells, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Dorothea Busse
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Claus Scheidereit
- Laboratory for Signal Transduction in Tumor Cells, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max Delbrueck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Mathematics and Computer Science, Free University Berlin, Germany
| |
Collapse
|
4
|
Argyris GA, Lluch Lafuente A, Tribastone M, Tschaikowski M, Vandin A. Reducing Boolean networks with backward equivalence. BMC Bioinformatics 2023; 24:212. [PMID: 37221494 DOI: 10.1186/s12859-023-05326-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/05/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Boolean Networks (BNs) are a popular dynamical model in biology where the state of each component is represented by a variable taking binary values that express, for instance, activation/deactivation or high/low concentrations. Unfortunately, these models suffer from the state space explosion, i.e., there are exponentially many states in the number of BN variables, which hampers their analysis. RESULTS We present Boolean Backward Equivalence (BBE), a novel reduction technique for BNs which collapses system variables that, if initialized with same value, maintain matching values in all states. A large-scale validation on 86 models from two online model repositories reveals that BBE is effective, since it is able to reduce more than 90% of the models. Furthermore, on such models we also show that BBE brings notable analysis speed-ups, both in terms of state space generation and steady-state analysis. In several cases, BBE allowed the analysis of models that were originally intractable due to the complexity. On two selected case studies, we show how one can tune the reduction power of BBE using model-specific information to preserve all dynamics of interest, and selectively exclude behavior that does not have biological relevance. CONCLUSIONS BBE complements existing reduction methods, preserving properties that other reduction methods fail to reproduce, and vice versa. BBE drops all and only the dynamics, including attractors, originating from states where BBE-equivalent variables have been initialized with different activation values The remaining part of the dynamics is preserved exactly, including the length of the preserved attractors, and their reachability from given initial conditions, without adding any spurious behaviours. Given that BBE is a model-to-model reduction technique, it can be combined with further reduction methods for BNs.
Collapse
Affiliation(s)
- Georgios A Argyris
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| | - Alberto Lluch Lafuente
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| | | | - Max Tschaikowski
- Department of Computer Science, University of Aalborg, Aalborg, Denmark
| | - Andrea Vandin
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark.
- Department of Excellence EMbeDS and Institute of Economics, Sant'Anna School for Advanced Studies, Pisa, Italy.
| |
Collapse
|
5
|
Su C, Pang J. Target Control of Asynchronous Boolean Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:707-719. [PMID: 34882560 DOI: 10.1109/tcbb.2021.3133608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
We study the target control of asynchronous Boolean networks, to identify interventions that can drive the dynamics of a given Boolean network from any initial state to the desired target attractor. Based on the application time, the control can be realised with three types of perturbations, including instantaneous, temporary and permanent perturbations. We develop efficient methods to compute the target control for a given target attractor with these three types of perturbations. We compare our methods with the stable motif-based control method on a variety of real-life biological networks to evaluate their performance. We show that our methods scale well for large Boolean networks and they are able to identify a rich set of solutions with a small number of perturbations.
Collapse
|
6
|
Basser-Ravitz E, Darbar A, Chifman J. Cyclic attractors of nonexpanding q-ary networks. J Math Biol 2022; 85:45. [PMID: 36203069 DOI: 10.1007/s00285-022-01796-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 06/28/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022]
Abstract
Discrete dynamical systems in which model components take on categorical values have been successfully applied to biological networks to study their global dynamic behavior. Boolean models in particular have been used extensively. However, multi-state models have also emerged as effective computational tools for the analysis of complex mechanisms underlying biological networks. Models in which variables assume more than two discrete states provide greater resolution, but this scheme introduces discontinuities. In particular, variables can increase or decrease by more than one unit in one time step. This can be corrected, without changing fixed points of the system, by applying an additional rule to each local activation function. On the other hand, if one is interested in cyclic attractors of their system, then this rule can potentially introduce new cyclic attractors that were not observed previously. This article makes some advancements in understanding the state space dynamics of multi-state network models with synchronous, sequential, or block-sequential update schedules and establishes conditions under which no new cyclic attractors are added to networks when the additional rule is applied. Our analytical results have the potential to be incorporated into modeling software and aid researchers in their analyses of biological multi-state networks.
Collapse
Affiliation(s)
| | | | - Julia Chifman
- Department of Mathematics and Statistics, American University, Washington, DC, USA.
| |
Collapse
|
7
|
Hérault L, Poplineau M, Remy E, Duprez E. Single Cell Transcriptomics to Understand HSC Heterogeneity and Its Evolution upon Aging. Cells 2022; 11:3125. [PMID: 36231086 PMCID: PMC9563410 DOI: 10.3390/cells11193125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/15/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Single-cell transcriptomic technologies enable the uncovering and characterization of cellular heterogeneity and pave the way for studies aiming at understanding the origin and consequences of it. The hematopoietic system is in essence a very well adapted model system to benefit from this technological advance because it is characterized by different cellular states. Each cellular state, and its interconnection, may be defined by a specific location in the global transcriptional landscape sustained by a complex regulatory network. This transcriptomic signature is not fixed and evolved over time to give rise to less efficient hematopoietic stem cells (HSC), leading to a well-documented hematopoietic aging. Here, we review the advance of single-cell transcriptomic approaches for the understanding of HSC heterogeneity to grasp HSC deregulations upon aging. We also discuss the new bioinformatics tools developed for the analysis of the resulting large and complex datasets. Finally, since hematopoiesis is driven by fine-tuned and complex networks that must be interconnected to each other, we highlight how mathematical modeling is beneficial for doing such interconnection between multilayered information and to predict how HSC behave while aging.
Collapse
Affiliation(s)
- Léonard Hérault
- I2M, CNRS, Aix Marseille University, 13009 Marseille, France
- Epigenetic Factors in Normal and Malignant Hematopoiesis Lab., CRCM, CNRS, INSERM, Institut Paoli Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Mathilde Poplineau
- Epigenetic Factors in Normal and Malignant Hematopoiesis Lab., CRCM, CNRS, INSERM, Institut Paoli Calmettes, Aix Marseille University, 13009 Marseille, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Elisabeth Remy
- I2M, CNRS, Aix Marseille University, 13009 Marseille, France
| | - Estelle Duprez
- Epigenetic Factors in Normal and Malignant Hematopoiesis Lab., CRCM, CNRS, INSERM, Institut Paoli Calmettes, Aix Marseille University, 13009 Marseille, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, 75013 Paris, France
| |
Collapse
|
8
|
Bhavani GS, Palanisamy A. SNAIL driven by a feed forward loop motif promotes TGF βinduced epithelial to mesenchymal transition. Biomed Phys Eng Express 2022; 8. [PMID: 35700712 DOI: 10.1088/2057-1976/ac7896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/14/2022] [Indexed: 11/12/2022]
Abstract
Epithelial to Mesenchymal Transition (EMT) plays an important role in tissue regeneration, embryonic development, and cancer metastasis. Several signaling pathways are known to regulate EMT, among which the modulation of TGFβ(Transforming Growth Factor-β) induced EMT is crucial in several cancer types. Several mathematical models were built to explore the role of core regulatory circuit of ZEB/miR-200, SNAIL/miR-34 double negative feedback loops in modulating TGFβinduced EMT. Different emergent behavior including tristability, irreversible switching, existence of hybrid EMT states were inferred though these models. Some studies have explored the role of TGFβreceptor activation, SMADs nucleocytoplasmic shuttling and complex formation. Recent experiments have revealed that MDM2 along with SMAD complex regulates SNAIL expression driven EMT. Encouraged by this, in the present study we developed a mathematical model for p53/MDM2 dependent TGFβinduced EMT regulation. Inclusion of p53 brings in an additional mechanistic perspective in exploring the EM transition. The network formulated comprises a C1FFL moderating SNAIL expression involving MDM2 and SMAD complex, which functions as a noise filter and persistent detector. The C1FFL was also observed to operate as a coincidence detector driving the SNAIL dependent downstream signaling into phenotypic switching decision. Systems modelling and analysis of the devised network, displayed interesting dynamic behavior, systems response to various inputs stimulus, providing a better understanding of p53/MDM2 dependent TGF-βinduced Epithelial to Mesenchymal Transition.
Collapse
|
9
|
Knapp AC, Sordo Vieira L, Laubenbacher R, Chifman J. SteadyCellPhenotype: a web-based tool for the modeling of biological networks with ternary logic. Bioinformatics 2022; 38:2369-2370. [PMID: 35179549 PMCID: PMC9004642 DOI: 10.1093/bioinformatics/btac097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/21/2021] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
SUMMARY We introduce SteadyCellPhenotype, a browser-based interface for the analysis of ternary biological networks. It includes tools for deterministically finding all steady states of a network, as well as the simulation and visualization of trajectories with publication quality graphics. Simulations allow us to approximate the size of the basin for attractors and deterministic simulations of trajectories nearby specified points allow us to explore the behavior of the system in that neighborhood. AVAILABILITY AND IMPLEMENTATION https://github.com/knappa/steadycellphenotype MIT License.
Collapse
Affiliation(s)
- Adam C Knapp
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA
- Department of Mathematics and Statistics, American University, Washington, DC 20016, USA
| | - Luis Sordo Vieira
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA
| | | | - Julia Chifman
- Department of Mathematics and Statistics, American University, Washington, DC 20016, USA
| |
Collapse
|
10
|
Stoll G, Naldi A, Noël V, Viara E, Barillot E, Kroemer G, Thieffry D, Calzone L. UPMaBoSS: A Novel Framework for Dynamic Cell Population Modeling. Front Mol Biosci 2022; 9:800152. [PMID: 35309516 PMCID: PMC8924294 DOI: 10.3389/fmolb.2022.800152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Mathematical modeling aims at understanding the effects of biological perturbations, suggesting ways to intervene and to reestablish proper cell functioning in diseases such as cancer or in autoimmune disorders. This is a difficult task for obvious reasons: the level of details needed to describe the intra-cellular processes involved, the numerous interactions between cells and cell types, and the complex dynamical properties of such populations where cells die, divide and interact constantly, to cite a few. Another important difficulty comes from the spatial distribution of these cells, their diffusion and motility. All of these aspects cannot be easily resolved in a unique mathematical model or with a unique formalism. To cope with some of these issues, we introduce here a novel framework, UPMaBoSS (for Update Population MaBoSS), dedicated to modeling dynamic populations of interacting cells. We rely on the preexisting tool MaBoSS, which enables probabilistic simulations of cellular networks. A novel software layer is added to account for cell interactions and population dynamics, but without considering the spatial dimension. This modeling approach can be seen as an intermediate step towards more complex spatial descriptions. We illustrate our methodology by means of a case study dealing with TNF-induced cell death. Interestingly, the simulation of cell population dynamics with UPMaBoSS reveals a mechanism of resistance triggered by TNF treatment. Relatively easy to encode, UPMaBoSS simulations require only moderate computational power and execution time. To ease the reproduction of simulations, we provide several Jupyter notebooks that can be accessed within the CoLoMoTo Docker image, which contains all software and models used for this study.
Collapse
Affiliation(s)
- Gautier Stoll
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Aurélien Naldi
- Institut de Biologie de L’ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
- Lifeware Group, Inria Saclay-Ile de France, Palaiseau, France
| | - Vincent Noël
- Institut Curie, PSL Research University, Paris, France
- INSERM U900, Paris, France
- MINES ParisTech, CBIO-Centre for Computational Biology, PSL Research University, Paris, France
| | | | - Emmanuel Barillot
- Institut Curie, PSL Research University, Paris, France
- INSERM U900, Paris, France
- MINES ParisTech, CBIO-Centre for Computational Biology, PSL Research University, Paris, France
| | - Guido Kroemer
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
- Pôle de Biologie, Hôpital européen Georges Pompidou, AP-HP, Paris, France
| | - Denis Thieffry
- Institut de Biologie de L’ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
- Lifeware Group, Inria Saclay-Ile de France, Palaiseau, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Paris, France
- INSERM U900, Paris, France
- MINES ParisTech, CBIO-Centre for Computational Biology, PSL Research University, Paris, France
- *Correspondence: Laurence Calzone,
| |
Collapse
|
11
|
Van Giang T, Akutsu T, Hiraishi K. An FVS-Based Approach to Attractor Detection in Asynchronous Random Boolean Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:806-818. [PMID: 33017287 DOI: 10.1109/tcbb.2020.3028862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Boolean networks (BNs)play a crucial role in modeling and analyzing biological systems. One of the central issues in the analysis of BNs is attractor detection, i.e., identification of all possible attractors. This problem becomes more challenging for large asynchronous random Boolean networks (ARBNs)because of the asynchronous and non-deterministic updating scheme. In this paper, we present and formally prove several relations between feedback vertex sets (FVSs)and dynamics of BNs. From these relations, we propose an FVS-based method for detecting attractors in ARBNs. Our approach relies on the principle of removing arcs in the state transition graph to get a candidate set and the reachability property to filter the candidate set. We formally prove the correctness of our method and show its efficiency by conducting experiments on real biological networks and randomly generated N- K networks. The obtained results are very promising since our method can handle large networks whose sizes are up to 101 without using any network reduction technique.
Collapse
|
12
|
Montagud A, Béal J, Tobalina L, Traynard P, Subramanian V, Szalai B, Alföldi R, Puskás L, Valencia A, Barillot E, Saez-Rodriguez J, Calzone L. Patient-specific Boolean models of signalling networks guide personalised treatments. eLife 2022; 11:e72626. [PMID: 35164900 PMCID: PMC9018074 DOI: 10.7554/elife.72626] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 01/27/2022] [Indexed: 11/22/2022] Open
Abstract
Prostate cancer is the second most occurring cancer in men worldwide. To better understand the mechanisms of tumorigenesis and possible treatment responses, we developed a mathematical model of prostate cancer which considers the major signalling pathways known to be deregulated. We personalised this Boolean model to molecular data to reflect the heterogeneity and specific response to perturbations of cancer patients. A total of 488 prostate samples were used to build patient-specific models and compared to available clinical data. Additionally, eight prostate cell line-specific models were built to validate our approach with dose-response data of several drugs. The effects of single and combined drugs were tested in these models under different growth conditions. We identified 15 actionable points of interventions in one cell line-specific model whose inactivation hinders tumorigenesis. To validate these results, we tested nine small molecule inhibitors of five of those putative targets and found a dose-dependent effect on four of them, notably those targeting HSP90 and PI3K. These results highlight the predictive power of our personalised Boolean models and illustrate how they can be used for precision oncology.
Collapse
Affiliation(s)
- Arnau Montagud
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3BarcelonaSpain
| | - Jonas Béal
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Luis Tobalina
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
| | - Pauline Traynard
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Vigneshwari Subramanian
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
| | - Bence Szalai
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
- Semmelweis University, Faculty of Medicine, Department of PhysiologyBudapestHungary
| | | | | | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC), Plaça Eusebi Güell, 1-3BarcelonaSpain
- ICREA, Pg. Lluís Companys 23BarcelonaSpain
| | - Emmanuel Barillot
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen UniversityAachenGermany
- Faculty of Medicine and Heidelberg University Hospital, Institute of Computational Biomedicine, Heidelberg UniversityHeidelbergGermany
| | - Laurence Calzone
- Institut Curie, PSL Research UniversityParisFrance
- INSERM, U900ParisFrance
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational BiologyParisFrance
| |
Collapse
|
13
|
Gondal MN, Chaudhary SU. Navigating Multi-Scale Cancer Systems Biology Towards Model-Driven Clinical Oncology and Its Applications in Personalized Therapeutics. Front Oncol 2021; 11:712505. [PMID: 34900668 PMCID: PMC8652070 DOI: 10.3389/fonc.2021.712505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022] Open
Abstract
Rapid advancements in high-throughput omics technologies and experimental protocols have led to the generation of vast amounts of scale-specific biomolecular data on cancer that now populates several online databases and resources. Cancer systems biology models built using this data have the potential to provide specific insights into complex multifactorial aberrations underpinning tumor initiation, development, and metastasis. Furthermore, the annotation of these single- and multi-scale models with patient data can additionally assist in designing personalized therapeutic interventions as well as aid in clinical decision-making. Here, we have systematically reviewed the emergence and evolution of (i) repositories with scale-specific and multi-scale biomolecular cancer data, (ii) systems biology models developed using this data, (iii) associated simulation software for the development of personalized cancer therapeutics, and (iv) translational attempts to pipeline multi-scale panomics data for data-driven in silico clinical oncology. The review concludes that the absence of a generic, zero-code, panomics-based multi-scale modeling pipeline and associated software framework, impedes the development and seamless deployment of personalized in silico multi-scale models in clinical settings.
Collapse
Affiliation(s)
- Mahnoor Naseer Gondal
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Safee Ullah Chaudhary
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
14
|
Thobe K, Konrath F, Chapuy B, Wolf J. Patient-Specific Modeling of Diffuse Large B-Cell Lymphoma. Biomedicines 2021; 9:biomedicines9111655. [PMID: 34829885 PMCID: PMC8615565 DOI: 10.3390/biomedicines9111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Personalized medicine aims to tailor treatment to patients based on their individual genetic or molecular background. Especially in diseases with a large molecular heterogeneity, such as diffuse large B-cell lymphoma (DLBCL), personalized medicine has the potential to improve outcome and/or to reduce resistance towards treatment. However, integration of patient-specific information into a computational model is challenging and has not been achieved for DLBCL. Here, we developed a computational model describing signaling pathways and expression of critical germinal center markers. The model integrates the regulatory mechanism of the signaling and gene expression network and covers more than 50 components, many carrying genetic lesions common in DLBCL. Using clinical and genomic data of 164 primary DLBCL patients, we implemented mutations, structural variants and copy number alterations as perturbations in the model using the CoLoMoTo notebook. Leveraging patient-specific genotypes and simulation of the expression of marker genes in specific germinal center conditions allows us to predict the consequence of the modeled pathways for each patient. Finally, besides modeling how genetic perturbations alter physiological signaling, we also predicted for each patient model the effect of rational inhibitors, such as Ibrutinib, that are currently discussed as possible DLBCL treatments, showing patient-dependent variations in effectiveness and synergies.
Collapse
Affiliation(s)
- Kirsten Thobe
- Mathematical Modelling of Cellular Processes, Max Delbrück Center for Molecular Medicine, 13125 Berlin-Buch, Germany; (K.T.); (F.K.)
| | - Fabian Konrath
- Mathematical Modelling of Cellular Processes, Max Delbrück Center for Molecular Medicine, 13125 Berlin-Buch, Germany; (K.T.); (F.K.)
| | - Björn Chapuy
- Department of Hematology and Medical Oncology, University of Göttingen, 37075 Göttingen, Germany;
- Department of Hematology, Oncology and Cancer Immunology, Berlin Medical Center Charité, 12203 Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max Delbrück Center for Molecular Medicine, 13125 Berlin-Buch, Germany; (K.T.); (F.K.)
- Department of Mathematics and Computer Science, Free University Berlin, Arnimallee 14, 14195 Berlin, Germany
- Correspondence:
| |
Collapse
|
15
|
Nuñez-Reza KJ, Naldi A, Sánchez-Jiménez A, Leon-Apodaca AV, Santana MA, Thomas-Chollier M, Thieffry D, Medina-Rivera A. Logical modelling of in vitro differentiation of human monocytes into dendritic cells unravels novel transcriptional regulatory interactions. Interface Focus 2021; 11:20200061. [PMID: 34123352 PMCID: PMC8193469 DOI: 10.1098/rsfs.2020.0061] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are the major specialized antigen-presenting cells, thereby connecting innate and adaptive immunity. Because of their role in establishing adaptive immunity, they constitute promising targets for immunotherapy. Monocytes can differentiate into DCs in vitro in the presence of colony-stimulating factor 2 (CSF2) and interleukin 4 (IL4), activating four signalling pathways (MAPK, JAK/STAT, NFKB and PI3K). However, the downstream transcriptional programme responsible for DC differentiation from monocytes (moDCs) remains unknown. By analysing the scientific literature on moDC differentiation, we established a preliminary logical model that helped us identify missing information regarding the activation of genes responsible for this differentiation, including missing targets for key transcription factors (TFs). Using ChIP-seq and RNA-seq data from the Blueprint consortium, we defined active and inactive promoters, together with differentially expressed genes in monocytes, moDCs and macrophages, which correspond to an alternative cell fate. We then used this functional genomic information to predict novel targets for previously identified TFs. By integrating this information, we refined our model and recapitulated the main established facts regarding moDC differentiation. Prospectively, the resulting model should be useful to develop novel immunotherapies targeting moDCs.
Collapse
Affiliation(s)
- Karen J Nuñez-Reza
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Aurélien Naldi
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Arantza Sánchez-Jiménez
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Ana V Leon-Apodaca
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - M Angélica Santana
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | - Morgane Thomas-Chollier
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Denis Thieffry
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| |
Collapse
|
16
|
Selvaggio G, Chaouiya C, Janody F. In Silico Logical Modelling to Uncover Cooperative Interactions in Cancer. Int J Mol Sci 2021; 22:ijms22094897. [PMID: 34063110 PMCID: PMC8125147 DOI: 10.3390/ijms22094897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022] Open
Abstract
The multistep development of cancer involves the cooperation between multiple molecular lesions, as well as complex interactions between cancer cells and the surrounding tumour microenvironment. The search for these synergistic interactions using experimental models made tremendous contributions to our understanding of oncogenesis. Yet, these approaches remain labour-intensive and challenging. To tackle such a hurdle, an integrative, multidisciplinary effort is required. In this article, we highlight the use of logical computational models, combined with experimental validations, as an effective approach to identify cooperative mechanisms and therapeutic strategies in the context of cancer biology. In silico models overcome limitations of reductionist approaches by capturing tumour complexity and by generating powerful testable hypotheses. We review representative examples of logical models reported in the literature and their validation. We then provide further analyses of our logical model of Epithelium to Mesenchymal Transition (EMT), searching for additional cooperative interactions involving inputs from the tumour microenvironment and gain of function mutations in NOTCH.
Collapse
Affiliation(s)
- Gianluca Selvaggio
- Fondazione the Microsoft Research—University of Trento Centre for Computational and Systems Biology (COSBI), Piazza Manifattura 1, 38068 Rovereto, Italy;
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
| | - Claudine Chaouiya
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
- CNRS, Centrale Marseille, I2M, Aix Marseille University, 13397 Marseille, France
- Correspondence: (C.C.); (F.J.)
| | - Florence Janody
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156 Oeiras, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Correspondence: (C.C.); (F.J.)
| |
Collapse
|
17
|
Béal J, Pantolini L, Noël V, Barillot E, Calzone L. Personalized logical models to investigate cancer response to BRAF treatments in melanomas and colorectal cancers. PLoS Comput Biol 2021; 17:e1007900. [PMID: 33507915 PMCID: PMC7872233 DOI: 10.1371/journal.pcbi.1007900] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 02/09/2021] [Accepted: 12/21/2020] [Indexed: 11/19/2022] Open
Abstract
The study of response to cancer treatments has benefited greatly from the contribution of different omics data but their interpretation is sometimes difficult. Some mathematical models based on prior biological knowledge of signaling pathways facilitate this interpretation but often require fitting of their parameters using perturbation data. We propose a more qualitative mechanistic approach, based on logical formalism and on the sole mapping and interpretation of omics data, and able to recover differences in sensitivity to gene inhibition without model training. This approach is showcased by the study of BRAF inhibition in patients with melanomas and colorectal cancers who experience significant differences in sensitivity despite similar omics profiles. We first gather information from literature and build a logical model summarizing the regulatory network of the mitogen-activated protein kinase (MAPK) pathway surrounding BRAF, with factors involved in the BRAF inhibition resistance mechanisms. The relevance of this model is verified by automatically assessing that it qualitatively reproduces response or resistance behaviors identified in the literature. Data from over 100 melanoma and colorectal cancer cell lines are then used to validate the model's ability to explain differences in sensitivity. This generic model is transformed into personalized cell line-specific logical models by integrating the omics information of the cell lines as constraints of the model. The use of mutations alone allows personalized models to correlate significantly with experimental sensitivities to BRAF inhibition, both from drug and CRISPR targeting, and even better with the joint use of mutations and RNA, supporting multi-omics mechanistic models. A comparison of these untrained models with learning approaches highlights similarities in interpretation and complementarity depending on the size of the datasets. This parsimonious pipeline, which can easily be extended to other biological questions, makes it possible to explore the mechanistic causes of the response to treatment, on an individualized basis.
Collapse
Affiliation(s)
- Jonas Béal
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Lorenzo Pantolini
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Vincent Noël
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Paris, France
- INSERM, U900, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| |
Collapse
|
18
|
Modelling of Immune Checkpoint Network Explains Synergistic Effects of Combined Immune Checkpoint Inhibitor Therapy and the Impact of Cytokines in Patient Response. Cancers (Basel) 2020; 12:cancers12123600. [PMID: 33276543 PMCID: PMC7761568 DOI: 10.3390/cancers12123600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/23/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The future of cancer immunotherapy relies on a combination of individually targeted therapies. However, a lot of experiments are needed to define the most effective combinations of drugs. A computational and modelling approach could help reduce the number of experiments and suggest optimal treatments to test. This article presents a logical model of T cell activation influenced by immune checkpoints, and explores the effect of these checkpoints, suggests mechanisms that would explain why some treatments might be better suited than others. The model includes not only programmed cell death protein 1 (PD1) and cytotoxic T-lymphocyte-associated protein 4 (CTL4) downstream pathways but also those of other immune checkpoints such as T cell immunoglobulin and ITIM (immunoreceptor tyrosine-based inhibition motif) domain (TIGIT), lymphocyte activation gene 3 (LAG3), T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), cluster of differentiation 226 (CD226), inducible T-cell costimulator (ICOS), and tumour necrosis factor receptors (TNFRs). Abstract After the success of the new generation of immune therapies, immune checkpoint receptors have become one important center of attention of molecular oncologists. The initial success and hopes of anti-programmed cell death protein 1 (anti-PD1) and anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA4) therapies have shown some limitations since a majority of patients have continued to show resistance. Other immune checkpoints have raised some interest and are under investigation, such as T cell immunoglobulin and ITIM (immunoreceptor tyrosine-based inhibition motif) domain (TIGIT), inducible T-cell costimulator (ICOS), and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), which appear as promising targets for immunotherapy. To explore their role and study possible synergetic effects of these different checkpoints, we have built a model of T cell receptor (TCR) regulation including not only PD1 and CTLA4, but also other well studied checkpoints (TIGIT, TIM3, lymphocyte activation gene 3 (LAG3), cluster of differentiation 226 (CD226), ICOS, and tumour necrosis factor receptors (TNFRs)) and simulated different aspects of T cell biology. Our model shows good correspondence with observations from available experimental studies of anti-PD1 and anti-CTLA4 therapies and suggest efficient combinations of immune checkpoint inhibitors (ICI). Among the possible candidates, TIGIT appears to be the most promising drug target in our model. The model predicts that signal transducer and activator of transcription 1 (STAT1)/STAT4-dependent pathways, activated by cytokines such as interleukin 12 (IL12) and interferon gamma (IFNG), could improve the effect of ICI therapy via upregulation of Tbet, suggesting that the effect of the cytokines related to STAT3/STAT1 activity is dependent on the balance between STAT1 and STAT3 downstream signalling.
Collapse
|
19
|
Pais RJ. Simulation of multiple microenvironments shows a pivot role of RPTPs on the control of Epithelial-to-Mesenchymal Transition. Biosystems 2020; 198:104268. [PMID: 33068671 DOI: 10.1016/j.biosystems.2020.104268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/22/2020] [Accepted: 10/01/2020] [Indexed: 10/23/2022]
Abstract
Epithelial-to-Mesenchymal Transition (EMT) is a natural and reversible process involved in embryogenesis, wound healing and thought to participate in the process of metastasis. Multiple signals from the microenvironment have been reported to drive EMT. However, the tight control of this process on physiological scenarios and how it is disrupted during cancer progression is not fully understood. Here, we analysed a regulatory network of EMT accounting for 10 key microenvironment signals focusing on the impact of two cell contact signals on the reversibility of EMT and the stability of resulting phenotypes. The analysis showed that the microenvironment is not enough for stabilizing Hybrid and Amoeboid-like phenotypes, requiring intracellular de-regulations as reported during cancer progression. Our simulations demonstrated that RPTP activation by cell contacts have the potential to inhibit the process of EMT and trigger its reversibility under tissue growth and chronic inflammation scenarios. Simulations also showed that hypoxia inhibits the capacity of RPTPs to control EMT. Our analysis further provided a theoretical explanation for the observed correlation between hypoxia and metastasis under chronic inflammation, and predicted that de-regulations in FAT4 signalling may promote Hybrid stabilization. Taken together, we propose a natural control mechanism of EMT that supports the idea that EMT is tightly regulated by the microenvironment.
Collapse
Affiliation(s)
- Ricardo Jorge Pais
- Centro de investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, Caparica, Portugal; BioenhancerSystems, London, UK.
| |
Collapse
|
20
|
Tokunaga R, Xiu J, Goldberg RM, Philip PA, Seeber A, Battaglin F, Arai H, Lo JH, Naseem M, Puccini A, Berger MD, Soni S, Zhang W, Chen S, Hwang JJ, Shields AF, Marshall JL, Baba H, Korn WM, Lenz HJ. The impact of ARID1A mutation on molecular characteristics in colorectal cancer. Eur J Cancer 2020; 140:119-129. [PMID: 33080474 DOI: 10.1016/j.ejca.2020.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/13/2020] [Accepted: 09/06/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND ARID1A is a key subunit of the SWItch/Sucrose Non-Fermentable (SWI/SNF) complex which regulates dynamic repositioning of nucleosomes to repair DNA damage. Only small pilot studies have evaluated the role of ARID1A mutation in colorectal cancer (CRC). The aim of the present study was to explore the potential impact of ARID1A mutation on clinicopathological and molecular characteristics in CRC. METHODS We used integrated data sets of 7978 CRC cases (one data set from a clinical laboratory improvement amendments [CLIA]-certified laboratory and three independent published data sets). The associations of ARID1A mutation with molecular characteristics including immune profile (the status of microsatellite instability [MSI], tumour mutational burden [TMB], programmed death ligand 1 [PD-L1] and estimated infiltrating immune cells), clinicopathological features and related pathways were analysed using next-generation sequencing, RNA sequencing and immunohistochemistry. RESULTS ARID1A mutant samples had more genomically unstable tumour features (MSI-high and TMB-high) and exhibited more characteristics of a T-cell-inflamed microenvironment (PD-L1 expression and high estimated infiltrating cytotoxic T lymphocytes [CTLs]) than ARID1A wild-type samples in the discovery and validation cohorts. Even ARID1A mutant samples without MSI-high status were TMB-high, had high levels of PD-L1 expression and high estimated infiltrating CTLs. ARID1A mutations were more common with right-sided primary and earlier stage tumours. ARID1A mutant tumours mainly had co-occurring gene mutations related to chromatin modifying, DNA repair, WNT signalling and epidermal growth factor receptor inhibitor resistance pathways, and ARID1A mutations strongly regulated DNA repair pathways. Key genes for chemotherapy/radiotherapy sensitivity were suppressed in ARID1A mutant samples. CONCLUSIONS Our findings may provide novel insights to develop individualised approaches for treatment of CRC based on ARID1A mutation status.
Collapse
Affiliation(s)
- Ryuma Tokunaga
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA; Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | | | | | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, USA
| | - Andreas Seeber
- Department of Haematology and Oncology, Innsbruck Medical University, Innsbruck, Austria
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Hiroyuki Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Jae Ho Lo
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Madiha Naseem
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Alberto Puccini
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Martin D Berger
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | | | - Jimmy J Hwang
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, USA
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, USA
| | - John L Marshall
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, USA
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - W Michael Korn
- Caris Life Sciences, Phoenix, USA; Division of Hematology, Oncology, University of California San Francisco, San Francisco, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| |
Collapse
|
21
|
Völkel G, Laban S, Fürstberger A, Kühlwein SD, Ikonomi N, Hoffmann TK, Brunner C, Neuberg DS, Gaidzik V, Döhner H, Kraus JM, Kestler HA. Analysis, identification and visualization of subgroups in genomics. Brief Bioinform 2020; 22:5909009. [PMID: 32954413 PMCID: PMC8138884 DOI: 10.1093/bib/bbaa217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/22/2022] Open
Abstract
Motivation Cancer is a complex and heterogeneous disease involving multiple somatic mutations that accumulate during its progression. In the past years, the wide availability of genomic data from patients’ samples opened new perspectives in the analysis of gene mutations and alterations. Hence, visualizing and further identifying genes mutated in massive sets of patients are nowadays a critical task that sheds light on more personalized intervention approaches. Results Here, we extensively review existing tools for visualization and analysis of alteration data. We compare different approaches to study mutual exclusivity and sample coverage in large-scale omics data. We complement our review with the standalone software AVAtar (‘analysis and visualization of alteration data’) that integrates diverse aspects known from different tools into a comprehensive platform. AVAtar supplements customizable alteration plots by a multi-objective evolutionary algorithm for subset identification and provides an innovative and user-friendly interface for the evaluation of concurrent solutions. A use case from personalized medicine demonstrates its unique features showing an application on vaccination target selection. Availability AVAtar is available at: https://github.com/sysbio-bioinf/avatar Contact hans.kestler@uni-ulm.de, phone: +49 (0) 731 500 24 500, fax: +49 (0) 731 500 24 502
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas K Hoffmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology, Head and Neck Surgery, Ulm University Medical Center, Germany
| | - Donna S Neuberg
- Department of Biostatistics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Verena Gaidzik
- Department of Internal Medicine III, Ulm University Medical Center, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Medical Center, Germany
| | | | | |
Collapse
|
22
|
Paulevé L, Kolčák J, Chatain T, Haar S. Reconciling qualitative, abstract, and scalable modeling of biological networks. Nat Commun 2020; 11:4256. [PMID: 32848126 PMCID: PMC7450094 DOI: 10.1038/s41467-020-18112-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/30/2020] [Indexed: 11/24/2022] Open
Abstract
Predicting biological systems' behaviors requires taking into account many molecular and genetic elements for which limited information is available past a global knowledge of their pairwise interactions. Logical modeling, notably with Boolean Networks (BNs), is a well-established approach that enables reasoning on the qualitative dynamics of networks. Several dynamical interpretations of BNs have been proposed. The synchronous and (fully) asynchronous ones are the most prominent, where the value of either all or only one component can change at each step. Here we prove that, besides being costly to analyze, these usual interpretations can preclude the prediction of certain behaviors observed in quantitative systems. We introduce an execution paradigm, the Most Permissive Boolean Networks (MPBNs), which offers the formal guarantee not to miss any behavior achievable by a quantitative model following the same logic. Moreover, MPBNs significantly reduce the complexity of dynamical analysis, enabling to model genome-scale networks.
Collapse
Affiliation(s)
- Loïc Paulevé
- Université Bordeaux, Bordeaux INP, CNRS, LaBRI, UMR5800, 351 cours de la Libération, Talence, 33400, France.
- LRI UMR8623, Université Paris-Sud, CNRS, Université Paris-Saclay, Bat 650 Ada Lovelace, Rue Raimond Castaing, Gif-sur-Yvette, 91190, France.
| | - Juri Kolčák
- Inria and LSV, CNRS (UMR 8643) and ENS Paris-Saclay, Université Paris-Saclay, 4 avenue des Sciences, Gif-sur-Yvette, 91190, France
| | - Thomas Chatain
- Inria and LSV, CNRS (UMR 8643) and ENS Paris-Saclay, Université Paris-Saclay, 4 avenue des Sciences, Gif-sur-Yvette, 91190, France
| | - Stefan Haar
- Inria and LSV, CNRS (UMR 8643) and ENS Paris-Saclay, Université Paris-Saclay, 4 avenue des Sciences, Gif-sur-Yvette, 91190, France
| |
Collapse
|
23
|
Simpson BS, Camacho N, Luxton HJ, Pye H, Finn R, Heavey S, Pitt J, Moore CM, Whitaker HC. Genetic alterations in the 3q26.31-32 locus confer an aggressive prostate cancer phenotype. Commun Biol 2020; 3:440. [PMID: 32796921 PMCID: PMC7429505 DOI: 10.1038/s42003-020-01175-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Large-scale genetic aberrations that underpin prostate cancer development and progression, such as copy-number alterations (CNAs), have been described but the consequences of specific changes in many identified loci is limited. Germline SNPs in the 3q26.31 locus are associated with aggressive prostate cancer, and is the location of NAALADL2, a gene overexpressed in aggressive disease. The closest gene to NAALADL2 is TBL1XR1, which is implicated in tumour development and progression. Using publicly-available cancer genomic data we report that NAALADL2 and TBL1XR1 gains/amplifications are more prevalent in aggressive sub-types of prostate cancer when compared to primary cohorts. In primary disease, gains/amplifications occurred in 15.99% (95% CI: 13.02–18.95) and 14.96% (95% CI: 12.08–17.84%) for NAALADL2 and TBL1XR1 respectively, increasing in frequency in higher Gleason grade and stage tumours. Gains/amplifications result in transcriptional changes and the development of a pro-proliferative and aggressive phenotype. These results support a pivotal role for copy-number gains in this genetic region. Benjamin Simpson et al. use publicly available cancer genomic data to investigate copy number changes at the 3q26.31–32 locus, which has been associated with aggressive prostate cancer based on single-nucleotide polymorphisms. They find that gains of NAALADL2 and TBL1XR1 in this locus are associated with more aggressive subtypes of prostate cancer and the transcription of pro-proliferative signalling processes.
Collapse
Affiliation(s)
- Benjamin S Simpson
- Molecular Diagnostics and Therapeutics Group, Research Department of Targeted Intervention, Division of Surgery & Interventional Science, University College London, London, UK
| | - Niedzica Camacho
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hayley J Luxton
- Molecular Diagnostics and Therapeutics Group, Research Department of Targeted Intervention, Division of Surgery & Interventional Science, University College London, London, UK
| | - Hayley Pye
- Molecular Diagnostics and Therapeutics Group, Research Department of Targeted Intervention, Division of Surgery & Interventional Science, University College London, London, UK
| | - Ron Finn
- Molecular Diagnostics and Therapeutics Group, Research Department of Targeted Intervention, Division of Surgery & Interventional Science, University College London, London, UK
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, Research Department of Targeted Intervention, Division of Surgery & Interventional Science, University College London, London, UK
| | - Jason Pitt
- Cancer Institute of Singapore, National University of Singapore, Singapore, Singapore
| | | | - Hayley C Whitaker
- Molecular Diagnostics and Therapeutics Group, Research Department of Targeted Intervention, Division of Surgery & Interventional Science, University College London, London, UK.
| |
Collapse
|
24
|
Zhou Y, Cheng X, Zhang F, Chen Q, Chen X, Shen Y, Lai C, Kota VG, Sun W, Huang Q, Yuan Y, Wang J, Lai M, Zhang D. Integrated multi-omics data analyses for exploring the co-occurring and mutually exclusive gene alteration events in colorectal cancer. Hum Mutat 2020; 41:1588-1599. [PMID: 32485022 DOI: 10.1002/humu.24059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/04/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022]
Abstract
Co-occurring and mutually exclusive gene alteration events are helpful for understanding carcinogenesis but systematic screening for such events is quite limited. We conducted pairwise screening tests to identify "hit pairs" in colorectal cancer (CRC) by utilizing the cross-omics data from The Cancer Genome Atlas (TCGA). Numerous hit pairs involving somatic mutations, copy number variations, and DNA methylation were found to occur nonrandomly in CRC, such as KRAS and HOXB6, SMAD4 and PMEPA1. Based on these hit pairs, we identified 32 synthetic lethal pairs and 7,527 co-occurring pairs relating to drug response. Our further biological experiments showed that the co-occurrence of mutant FCGBP and NUDT12 silencing (or mutant TMC3 and RPS6KA6 silencing) with small interfering RNA reduced cell viability. Moreover, novel hit pairs could influence prognosis. The patients who carried concurrent mutations of IRF5 and NEFH, SYNE1 and TTN, or MUC16 and NEFH had worse survival outcomes. Particularly, the presence of mutant SYNE1 and TTN pair not only affects prognosis, but also is related to CRC patients' response to drug treatment. Our "hit pair" genes may provide insights into colorectal carcinogenesis and help open new avenues for CRC therapy.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Pathology, and Department of Medical Oncology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoqing Cheng
- Department of Pathology, and Department of Medical Oncology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fenglan Zhang
- Department of Pathology, and Department of Medical Oncology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qingqing Chen
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyu Chen
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yaojia Shen
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chong Lai
- Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Vishnu G Kota
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenjie Sun
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiong Huang
- The Core Facilities, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Yuan
- Department of Medical Oncology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Chinese National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jin Wang
- Department of Pathology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Maode Lai
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dandan Zhang
- Department of Pathology, and Department of Medical Oncology of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Klarner H, Heinitz F, Nee S, Siebert H. Basins of Attraction, Commitment Sets, and Phenotypes of Boolean Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1115-1124. [PMID: 30575543 DOI: 10.1109/tcbb.2018.2879097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The attractors of Boolean networks and their basins have been shown to be highly relevant for model validation and predictive modeling, e.g., in systems biology. Yet, there are currently very few tools available that are able to compute and visualize not only attractors but also their basins. In the realm of asynchronous, non-deterministic modeling not only is the repertoire of software even more limited, but also the formal notions for basins of attraction are often lacking. In this setting, the difficulty both for theory and computation arises from the fact that states may be elements of several distinct basins. In this paper, we address this topic by partitioning the state space into sets that are committed to the same attractors. These commitment sets can easily be generalized to sets that are equivalent w.r.t. the long-term behaviors of pre-selected nodes which leads us to the notions of markers and phenotypes which we illustrate in a case study on bladder tumorigenesis. For every concept, we propose equivalent CTL model checking queries and an extension of the state of the art model checking software NuSMV is made available that is capable of computing the respective sets. All notions are fully integrated as three new modules in our Python package PyBoolNet, including functions for visualizing the basins, commitment sets, and phenotypes as quotient graphs and pie charts.
Collapse
|
26
|
Ho AS, Ochoa A, Jayakumaran G, Zehir A, Valero Mayor C, Tepe J, Makarov V, Dalin MG, He J, Bailey M, Montesion M, Ross JS, Miller VA, Chan L, Ganly I, Dogan S, Katabi N, Tsipouras P, Ha P, Agrawal N, Solit DB, Futreal PA, El Naggar AK, Reis-Filho JS, Weigelt B, Ho AL, Schultz N, Chan TA, Morris LG. Genetic hallmarks of recurrent/metastatic adenoid cystic carcinoma. J Clin Invest 2020; 129:4276-4289. [PMID: 31483290 DOI: 10.1172/jci128227] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/09/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUNDAdenoid cystic carcinoma (ACC) is a rare malignancy arising in salivary glands and other sites, characterized by high rates of relapse and distant spread. Recurrent/metastatic (R/M) ACCs are generally incurable, due to a lack of active systemic therapies. To improve outcomes, deeper understanding of genetic alterations and vulnerabilities in R/M tumors is needed.METHODSAn integrated genomic analysis of 1,045 ACCs (177 primary, 868 R/M) was performed to identify alterations associated with advanced and metastatic tumors. Intratumoral genetic heterogeneity, germline mutations, and therapeutic actionability were assessed.RESULTSCompared with primary tumors, R/M tumors were enriched for alterations in key Notch (NOTCH1, 26.3% vs. 8.5%; NOTCH2, 4.6% vs. 2.3%; NOTCH3, 5.7% vs. 2.3%; NOTCH4, 3.6% vs. 0.6%) and chromatin-remodeling (KDM6A, 15.2% vs. 3.4%; KMT2C/MLL3, 14.3% vs. 4.0%; ARID1B, 14.1% vs. 4.0%) genes. TERT promoter mutations (13.1% of R/M cases) were mutually exclusive with both NOTCH1 mutations (q = 3.3 × 10-4) and MYB/MYBL1 fusions (q = 5.6 × 10-3), suggesting discrete, alternative mechanisms of tumorigenesis. This network of alterations defined 4 distinct ACC subgroups: MYB+NOTCH1+, MYB+/other, MYBWTNOTCH1+, and MYBWTTERT+. Despite low mutational load, we identified numerous samples with marked intratumoral genetic heterogeneity, including branching evolution across multiregion sequencing.CONCLUSIONThese observations collectively redefine the molecular underpinnings of ACC progression and identify further targets for precision therapies.FUNDINGAdenoid Cystic Carcinoma Research Foundation, Pershing Square Sohn Cancer Research grant, the PaineWebber Chair, Stand Up 2 Cancer, NIH R01 CA205426, the STARR Cancer Consortium, NCI R35 CA232097, the Frederick Adler Chair, Cycle for Survival, the Jayme Flowers Fund, The Sebastian Nativo Fund, NIH K08 DE024774 and R01 DE027738, and MSKCC through NIH/NCI Cancer Center Support Grant (P30 CA008748).
Collapse
Affiliation(s)
- Allen S Ho
- Department of Surgery and.,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Angelica Ochoa
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology
| | | | | | | | - Justin Tepe
- Head and Neck Service, Department of Surgery, and
| | - Vladimir Makarov
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Martin G Dalin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Jie He
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Mark Bailey
- Foundation Medicine, Cambridge, Massachusetts, USA
| | | | | | | | - Lindsay Chan
- Foundation Medicine, Cambridge, Massachusetts, USA
| | - Ian Ganly
- Head and Neck Service, Department of Surgery, and
| | | | | | - Petros Tsipouras
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Patrick Ha
- Department of Otolaryngology-Head and Neck Surgery, UCSF, San Francisco, California, USA
| | - Nishant Agrawal
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - David B Solit
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology.,Head and Neck Service, Department of Surgery, and.,Department of Medicine
| | | | - Adel K El Naggar
- Department of Pathology, University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas, USA
| | | | - Britta Weigelt
- Experimental Pathology Service, MSKCC, New York, New York, USA
| | | | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA.,Department of Radiation Oncology, and.,Immunogenomics and Precision Oncology Platform, MSKCC, New York, New York, USA
| | - Luc Gt Morris
- Head and Neck Service, Department of Surgery, and.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA.,Immunogenomics and Precision Oncology Platform, MSKCC, New York, New York, USA
| |
Collapse
|
27
|
Stalidzans E, Zanin M, Tieri P, Castiglione F, Polster A, Scheiner S, Pahle J, Stres B, List M, Baumbach J, Lautizi M, Van Steen K, Schmidt HH. Mechanistic Modeling and Multiscale Applications for Precision Medicine: Theory and Practice. NETWORK AND SYSTEMS MEDICINE 2020. [DOI: 10.1089/nsm.2020.0002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Egils Stalidzans
- Computational Systems Biology Group, University of Latvia, Riga, Latvia
- Latvian Biomedical Reasearch and Study Centre, Riga, Latvia
| | - Massimiliano Zanin
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Spain
| | - Paolo Tieri
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | - Filippo Castiglione
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | | | - Stefan Scheiner
- Institute for Mechanics of Materials and Structures, Vienna University of Technology, Vienna, Austria
| | - Jürgen Pahle
- BioQuant, Heidelberg University, Heidelberg, Germany
| | - Blaž Stres
- Department of Animal Science, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Civil and Geodetic Engineering, University of Ljubljana, Ljubljana, Slovenia
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Markus List
- Big Data in BioMedicine Research Group, Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Manuela Lautizi
- Computational Systems Medicine Research Group, Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Kristel Van Steen
- BIO-Systems Genetics, GIGA-R, University of Liège, Liège, Belgium
- BIO3—Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Harald H.H.W. Schmidt
- Department of Pharmacology and Personalised Medicine, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
28
|
Montagud A, Traynard P, Martignetti L, Bonnet E, Barillot E, Zinovyev A, Calzone L. Conceptual and computational framework for logical modelling of biological networks deregulated in diseases. Brief Bioinform 2020; 20:1238-1249. [PMID: 29237040 DOI: 10.1093/bib/bbx163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/24/2017] [Indexed: 01/02/2023] Open
Abstract
Mathematical models can serve as a tool to formalize biological knowledge from diverse sources, to investigate biological questions in a formal way, to test experimental hypotheses, to predict the effect of perturbations and to identify underlying mechanisms. We present a pipeline of computational tools that performs a series of analyses to explore a logical model's properties. A logical model of initiation of the metastatic process in cancer is used as a transversal example. We start by analysing the structure of the interaction network constructed from the literature or existing databases. Next, we show how to translate this network into a mathematical object, specifically a logical model, and how robustness analyses can be applied to it. We explore the visualization of the stable states, defined as specific attractors of the model, and match them to cellular fates or biological read-outs. With the different tools we present here, we explain how to assign to each solution of the model a probability and how to identify genetic interactions using mutant phenotype probabilities. Finally, we connect the model to relevant experimental data: we present how some data analyses can direct the construction of the network, and how the solutions of a mathematical model can also be compared with experimental data, with a particular focus on high-throughput data in cancer biology. A step-by-step tutorial is provided as a Supplementary Material and all models, tools and scripts are provided on an accompanying website: https://github.com/sysbio-curie/Logical_modelling_pipeline.
Collapse
|
29
|
Borriello E, Walker SI, Laubichler MD. Cell phenotypes as macrostates of the GRN dynamics. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2020; 334:213-224. [DOI: 10.1002/jez.b.22938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 01/04/2023]
Affiliation(s)
- Enrico Borriello
- ASU‐SFI Center for Biosocial Complex SystemsArizona State UniversityTempe Arizona
| | - Sara I. Walker
- ASU‐SFI Center for Biosocial Complex SystemsArizona State UniversityTempe Arizona
- Beyond Center for Fundamental Concepts in ScienceArizona State UniversityTempe Arizona
- School of Earth and Space ExplorationArizona State UniversityTempe Arizona
- Blue Marble Space Institute of ScienceSeattle Washington
| | - Manfred D. Laubichler
- ASU‐SFI Center for Biosocial Complex SystemsArizona State UniversityTempe Arizona
- Santa Fe InstituteSanta Fe New Mexico
- Marine Biological LaboratoryWoods Hole Massachusetts
- School of Life SciencesArizona State UniversityTempe Arizona
| |
Collapse
|
30
|
Sordo Vieira L, Laubenbacher RC, Murrugarra D. Control of Intracellular Molecular Networks Using Algebraic Methods. Bull Math Biol 2019; 82:2. [PMID: 31919596 DOI: 10.1007/s11538-019-00679-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/02/2019] [Indexed: 10/25/2022]
Abstract
Many problems in biology and medicine have a control component. Often, the goal might be to modify intracellular networks, such as gene regulatory networks or signaling networks, in order for cells to achieve a certain phenotype, what happens in cancer. If the network is represented by a mathematical model for which mathematical control approaches are available, such as systems of ordinary differential equations, then this problem might be solved systematically. Such approaches are available for some other model types, such as Boolean networks, where structure-based approaches have been developed, as well as stable motif techniques. However, increasingly many published discrete models are mixed-state or multistate, that is, some or all variables have more than two states, and thus the development of control strategies for multistate networks is needed. This paper presents a control approach broadly applicable to general multistate models based on encoding them as polynomial dynamical systems over a finite algebraic state set, and using computational algebra for finding appropriate intervention strategies. To demonstrate the feasibility and applicability of this method, we apply it to a recently developed multistate intracellular model of E2F-mediated bladder cancerous growth and to a model linking intracellular iron metabolism and oncogenic pathways. The control strategies identified for these published models are novel in some cases and represent new hypotheses, or are supported by the literature in others as potential drug targets. Our Macaulay2 scripts to find control strategies are publicly available through GitHub at https://github.com/luissv7/multistatepdscontrol.
Collapse
Affiliation(s)
- Luis Sordo Vieira
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA
| | - Reinhard C Laubenbacher
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT, 06032, USA.,Center for Quantitative Medicine, UConn Health, Farmington, CT, 06032, USA
| | - David Murrugarra
- Department of Mathematics, University of Kentucky, Lexington, KY, 40506, USA.
| |
Collapse
|
31
|
Ho AS, Ochoa A, Jayakumaran G, Zehir A, Valero Mayor C, Tepe J, Makarov V, Dalin MG, He J, Bailey M, Montesion M, Ross JS, Miller VA, Chan L, Ganly I, Dogan S, Katabi N, Tsipouras P, Ha P, Agrawal N, Solit DB, Futreal PA, El Naggar AK, Reis-Filho JS, Weigelt B, Ho AL, Schultz N, Chan TA, Morris LG. Genetic hallmarks of recurrent/metastatic adenoid cystic carcinoma. J Clin Invest 2019. [DOI: 10.1172/jci128227 pmid:314832902019-10-01]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
32
|
Mandon H, Su C, Pang J, Paul S, Haar S, Pauleve L. Algorithms for the Sequential Reprogramming of Boolean Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:1610-1619. [PMID: 31056515 DOI: 10.1109/tcbb.2019.2914383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cellular reprogramming, a technique that opens huge opportunities in modern and regenerative medicine, heavily relies on identifying key genes to perturb. Most of the existing computational methods for controlling which attractor (steady state) the cell will reach focus on finding mutations to apply to the initial state. However, it has been shown, and is proved in this article, that waiting between perturbations so that the update dynamics of the system prepares the ground, allows for new reprogramming strategies. To identify such sequential perturbations, we consider a qualitative model of regulatory networks, and rely on Binary Decision Diagrams to model their dynamics and the putative perturbations. Our method establishes a set identification of sequential perturbations, whether permanent (mutations) or only temporary, to achieve the existential or inevitable reachability of an arbitrary state of the system. We apply an implementation for temporary perturbations on models from the literature, illustrating that we are able to derive sequential perturbations to achieve trans-differentiation.
Collapse
|
33
|
Bag AK, Mandloi S, Jarmalavicius S, Mondal S, Kumar K, Mandal C, Walden P, Chakrabarti S, Mandal C. Connecting signaling and metabolic pathways in EGF receptor-mediated oncogenesis of glioblastoma. PLoS Comput Biol 2019; 15:e1007090. [PMID: 31386654 PMCID: PMC6684045 DOI: 10.1371/journal.pcbi.1007090] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 05/13/2019] [Indexed: 12/21/2022] Open
Abstract
As malignant transformation requires synchronization of growth-driving signaling (S) and metabolic (M) pathways, defining cancer-specific S-M interconnected networks (SMINs) could lead to better understanding of oncogenic processes. In a systems-biology approach, we developed a mathematical model for SMINs in mutated EGF receptor (EGFRvIII) compared to wild-type EGF receptor (EGFRwt) expressing glioblastoma multiforme (GBM). Starting with experimentally validated human protein-protein interactome data for S-M pathways, and incorporating proteomic data for EGFRvIII and EGFRwt GBM cells and patient transcriptomic data, we designed a dynamic model for EGFR-driven GBM-specific information flow. Key nodes and paths identified by in silico perturbation were validated experimentally when inhibition of signaling pathway proteins altered expression of metabolic proteins as predicted by the model. This demonstrated capacity of the model to identify unknown connections between signaling and metabolic pathways, explain the robustness of oncogenic SMINs, predict drug escape, and assist identification of drug targets and the development of combination therapies. Complex and highly dynamic interconnected networks allow cancer to take different routes and circumvent chemotherapy. Therefore, understanding these context-specific networks and their dynamics of molecular interactions driven by different oncogenic signaling and metabolic pathways is very much needed to predict drug targets and the effect of therapeutics. We incorporated high-throughput transcriptome and proteome data into mathematical models to deduce properties of cancer cells through systems biology approach. Here we report the development, testing and validation of an integrated systems biology model of information flow between signaling and metabolic pathways to understand the regulation of the interconnection between them in cancer. Our model efficiently identified unique connections and key nodes important in signaling-metabolic information flow. We predicted some potential novel targets before performing actual drug tests. We have successfully applied this model to identify the interconnections altered in the constitutive signaling of the mutated EGFR by comparing EGF-dependent and wild-type EGFR signaling in glioblastoma multiforme.
Collapse
Affiliation(s)
- Arup K. Bag
- Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Sapan Mandloi
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Saulius Jarmalavicius
- Department of Dermatology, Venerology and Allergology, Charité– Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susmita Mondal
- Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Krishna Kumar
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Kolkata, India
| | - Chhabinath Mandal
- National Institute of Pharmaceutical Education and Research, Kolkata, India
| | - Peter Walden
- Department of Dermatology, Venerology and Allergology, Charité– Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- * E-mail: (PW); , (SC); , (CM)
| | - Saikat Chakrabarti
- Structural Biology and Bioinformatics Division, Indian Institute of Chemical Biology, Kolkata, India
- * E-mail: (PW); , (SC); , (CM)
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, Indian Institute of Chemical Biology, Kolkata, India
- * E-mail: (PW); , (SC); , (CM)
| |
Collapse
|
34
|
Deng Y, Luo S, Deng C, Luo T, Yin W, Zhang H, Zhang Y, Zhang X, Lan Y, Ping Y, Xiao Y, Li X. Identifying mutual exclusivity across cancer genomes: computational approaches to discover genetic interaction and reveal tumor vulnerability. Brief Bioinform 2019; 20:254-266. [PMID: 28968730 DOI: 10.1093/bib/bbx109] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Indexed: 02/06/2023] Open
Abstract
Systematic sequencing of cancer genomes has revealed prevalent heterogeneity, with patients harboring various combinatorial patterns of genetic alteration. In particular, a phenomenon that a group of genes exhibits mutually exclusive patterns has been widespread across cancers, covering a broad spectrum of crucial cancer pathways. Recently, there is considerable evidence showing that, mutual exclusivity reflects alternative functions in tumor initiation and progression, or suggests adverse effects of their concurrence. Given its importance, numerous computational approaches have been proposed to study mutual exclusivity using genomic profiles alone, or by integrating networks and phenotypes. Some of them have been routinely used to explore genetic associations, which lead to a deeper understanding of carcinogenic mechanisms and reveals unexpected tumor vulnerabilities. Here, we present an overview of mutual exclusivity from the perspective of cancer genome. We describe the common hypothesis underlying mutual exclusivity, summarize the strategies for the identification of significant mutually exclusive patterns, compare the performance of representative algorithms from simulated data sets and discuss their common confounders.
Collapse
Affiliation(s)
- Yulan Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shangyi Luo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chunyu Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tao Luo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenkang Yin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyi Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanyan Ping
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
35
|
Baudin A, Paul S, Su C, Pang J. Controlling large Boolean networks with single-step perturbations. Bioinformatics 2019; 35:i558-i567. [PMID: 31510648 PMCID: PMC6612811 DOI: 10.1093/bioinformatics/btz371] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Motivation The control of Boolean networks has traditionally focussed on strategies where the perturbations are applied to the nodes of the network for an extended period of time. In this work, we study if and how a Boolean network can be controlled by perturbing a minimal set of nodes for a single-step and letting the system evolve afterwards according to its original dynamics. More precisely, given a Boolean network (BN), we compute a minimal subset Cmin of the nodes such that BN can be driven from any initial state in an attractor to another ‘desired’ attractor by perturbing some or all of the nodes of Cmin for a single-step. Such kind of control is attractive for biological systems because they are less time consuming than the traditional strategies for control while also being financially more viable. However, due to the phenomenon of state-space explosion, computing such a minimal subset is computationally inefficient and an approach that deals with the entire network in one-go, does not scale well for large networks. Results We develop a ‘divide-and-conquer’ approach by decomposing the network into smaller partitions, computing the minimal control on the projection of the attractors to these partitions and then composing the results to obtain Cmin for the whole network. We implement our method and test it on various real-life biological networks to demonstrate its applicability and efficiency. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Alexis Baudin
- Department of Computer Science, École Normale Supérieure Paris-Saclay, Cachan, France
| | - Soumya Paul
- Faculty of Science, Technology and Communication, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cui Su
- Interdisciplinary Centre for Security, Reliability and Trust, University of Luxembourg, Luxembourg, Luxembourg
| | - Jun Pang
- Faculty of Science, Technology and Communication, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,Interdisciplinary Centre for Security, Reliability and Trust, University of Luxembourg, Luxembourg, Luxembourg
| |
Collapse
|
36
|
Wery M, Dameron O, Nicolas J, Remy E, Siegel A. Formalizing and enriching phenotype signatures using Boolean networks. J Theor Biol 2019; 467:66-79. [DOI: 10.1016/j.jtbi.2019.01.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/30/2018] [Accepted: 01/08/2019] [Indexed: 01/12/2023]
|
37
|
Béal J, Montagud A, Traynard P, Barillot E, Calzone L. Personalization of Logical Models With Multi-Omics Data Allows Clinical Stratification of Patients. Front Physiol 2019; 9:1965. [PMID: 30733688 PMCID: PMC6353844 DOI: 10.3389/fphys.2018.01965] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 12/31/2018] [Indexed: 12/26/2022] Open
Abstract
Logical models of cancer pathways are typically built by mining the literature for relevant experimental observations. They are usually generic as they apply for large cohorts of individuals. As a consequence, they generally do not capture the heterogeneity of patient tumors and their therapeutic responses. We present here a novel framework, referred to as PROFILE, to tailor logical models to a particular biological sample such as a patient tumor. This methodology permits to compare the model simulations to individual clinical data, i.e., survival time. Our approach focuses on integrating mutation data, copy number alterations (CNA), and expression data (transcriptomics or proteomics) to logical models. These data need first to be either binarized or set between 0 and 1, and can then be incorporated in the logical model by modifying the activity of the node, the initial conditions or the state transition rates. The use of MaBoSS, a tool based on Monte-Carlo kinetic algorithm to perform stochastic simulations on logical models results in model state probabilities, and allows for a semi-quantitative study of the model phenotypes and perturbations. As a proof of concept, we use a published generic model of cancer signaling pathways and molecular data from METABRIC breast cancer patients. For this example, we test several combinations of data incorporation and discuss that, with these data, the most comprehensive patient-specific cancer models are obtained by modifying the nodes' activity of the model with mutations, in combination or not with CNA data, and altering the transition rates with RNA expression. We conclude that these model simulations show good correlation with clinical data such as patients' Nottingham prognostic index (NPI) subgrouping and survival time. We observe that two highly relevant cancer phenotypes derived from personalized models, Proliferation and Apoptosis, are biologically consistent prognostic factors: patients with both high proliferation and low apoptosis have the worst survival rate, and conversely. Our approach aims to combine the mechanistic insights of logical modeling with multi-omics data integration to provide patient-relevant models. This work leads to the use of logical modeling for precision medicine and will eventually facilitate the choice of patient-specific drug treatments by physicians.
Collapse
Affiliation(s)
| | | | | | - Emmanuel Barillot
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| |
Collapse
|
38
|
Apollo A, Ortenzi V, Scatena C, Zavaglia K, Aretini P, Lessi F, Franceschi S, Tomei S, Sepich CA, Viacava P, Mazzanti CM, Naccarato AG. Molecular characterization of low grade and high grade bladder cancer. PLoS One 2019; 14:e0210635. [PMID: 30650148 PMCID: PMC6334926 DOI: 10.1371/journal.pone.0210635] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/29/2018] [Indexed: 12/19/2022] Open
Abstract
Background Bladder cancer (BC) is the 9th most common cancer diagnosis worldwide. Low grade (LG) represents 70% of all BCs, characterized by recurrence and rare ability (10–15%) to progress to high grade (HG) and invade. The remaining 30% is high grade (HG), fast invasive BC, which is resistant to therapy. Identifying biomarkers for predicting those tumors able to progress is a key goal for patient outcome improvement. This study focuses on the most promising prognostic markers. Materials and methods TP53 and FGFR3 mutational status, Survivin, CK19, CK20, E-cadherin and CD44 gene expression analysis were performed on 66 BCs. Results Survivin was found associated to tumor grade (p<0.05). Moreover, Survivin correlated with CD44 in TP53 wild type (p = 0.0242) and FGFR3 wild type (p = 0.0036) tumors. In particular the Survivin-CD44 correlation was associated to HG FGFR3 wild type BCs (p = 0.0045). Unsupervised hierarchical clustering based on gene expression data identified four distinct molecular groups reflecting the patient histology (p = 0.038). Conclusion We suggest Survivin, both as a biomarker associated to G3 BCs but negatively related to TP53 mutational status, and as a potential novel therapeutic target.
Collapse
Affiliation(s)
- Alessandro Apollo
- Genetic Unit of Biology Department, University of Pisa, Pisa, Italy
- * E-mail:
| | - Valerio Ortenzi
- Department of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Cristian Scatena
- Department of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Katia Zavaglia
- Department of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Paolo Aretini
- Section of Cancer Genomics, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Francesca Lessi
- Section of Cancer Genomics, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Sara Franceschi
- Section of Cancer Genomics, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Sara Tomei
- Omics Core and Biorepository, Sidra Medicine, Doha, Qatar
| | | | - Paolo Viacava
- Division of Pathology, Hospital of Livorno, Livorno, Italy
| | | | | |
Collapse
|
39
|
Deng Y, Luo S, Zhang X, Zou C, Yuan H, Liao G, Xu L, Deng C, Lan Y, Zhao T, Gao X, Xiao Y, Li X. A pan-cancer atlas of cancer hallmark-associated candidate driver lncRNAs. Mol Oncol 2018; 12:1980-2005. [PMID: 30216655 PMCID: PMC6210054 DOI: 10.1002/1878-0261.12381] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 07/21/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
Substantial cancer genome sequencing efforts have discovered many important driver genes contributing to tumorigenesis. However, very little is known about the genetic alterations of long non‐coding RNAs (lncRNAs) in cancer. Thus, there is a need for systematic surveys of driver lncRNAs. Through integrative analysis of 5918 tumors across 11 cancer types, we revealed that lncRNAs have undergone dramatic genomic alterations, many of which are mutually exclusive with well‐known cancer genes. Using the hypothesis of functional redundancy of mutual exclusivity, we developed a computational framework to identify driver lncRNAs associated with different cancer hallmarks. Applying it to pan‐cancer data, we identified 378 candidate driver lncRNAs whose genomic features highly resemble the known cancer driver genes (e.g. high conservation and early replication). We further validated the candidate driver lncRNAs involved in ‘Tissue Invasion and Metastasis’ in lung adenocarcinoma and breast cancer, and also highlighted their potential roles in improving clinical outcomes. In summary, we have generated a comprehensive landscape of cancer candidate driver lncRNAs that could act as a starting point for future functional explorations, as well as the identification of biomarkers and lncRNA‐based target therapy.
Collapse
Affiliation(s)
- Yulan Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Shangyi Luo
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Chaoxia Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, China
| | - Huating Yuan
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Gaoming Liao
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Liwen Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Chunyu Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Yujia Lan
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Tingting Zhao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, China.,Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, China.,Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, China
| |
Collapse
|
40
|
Mendes ND, Henriques R, Remy E, Carneiro J, Monteiro PT, Chaouiya C. Estimating Attractor Reachability in Asynchronous Logical Models. Front Physiol 2018; 9:1161. [PMID: 30245634 PMCID: PMC6137237 DOI: 10.3389/fphys.2018.01161] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/02/2018] [Indexed: 12/12/2022] Open
Abstract
Logical models are well-suited to capture salient dynamical properties of regulatory networks. For networks controlling cell fate decisions, cell fates are associated with model attractors (stable states or cyclic attractors) whose identification and reachability properties are particularly relevant. While synchronous updates assume unlikely instantaneous or identical rates associated with component changes, the consideration of asynchronous updates is more realistic but, for large models, may hinder the analysis of the resulting non-deterministic concurrent dynamics. This complexity hampers the study of asymptotical behaviors, and most existing approaches suffer from efficiency bottlenecks, being generally unable to handle cyclical attractors and quantify attractor reachability. Here, we propose two algorithms providing probability estimates of attractor reachability in asynchronous dynamics. The first algorithm, named Firefront, exhaustively explores the state space from an initial state, and provides quasi-exact evaluations of the reachability probabilities of model attractors. The algorithm progresses in breadth, propagating the probabilities of each encountered state to its successors. Second, Avatar is an adapted Monte Carlo approach, better suited for models with large and intertwined transient and terminal cycles. Avatar iteratively explores the state space by randomly selecting trajectories and by using these random walks to estimate the likelihood of reaching an attractor. Unlike Monte Carlo simulations, Avatar is equipped to avoid getting trapped in transient cycles and to identify cyclic attractors. Firefront and Avatar are validated and compared to related methods, using as test cases logical models of synthetic and biological networks. Both algorithms are implemented as new functionalities of GINsim 3.0, a well-established software tool for logical modeling, providing executable GUI, Java API, and scripting facilities.
Collapse
Affiliation(s)
| | - Rui Henriques
- Department of Computer Science and Engineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Engenharia de Sistemas e Computadores Investigação e Desenvolvimento, Lisbon, Portugal
| | - Elisabeth Remy
- Aix Marseille University, CNRS, Centrale Marseille, I2M UMR 7373, Marseille, France
| | | | - Pedro T Monteiro
- Department of Computer Science and Engineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Engenharia de Sistemas e Computadores Investigação e Desenvolvimento, Lisbon, Portugal
| | | |
Collapse
|
41
|
Calzone L, Barillot E, Zinovyev A. Logical versus kinetic modeling of biological networks: applications in cancer research. Curr Opin Chem Eng 2018. [DOI: 10.1016/j.coche.2018.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
42
|
Naldi A, Hernandez C, Abou-Jaoudé W, Monteiro PT, Chaouiya C, Thieffry D. Logical Modeling and Analysis of Cellular Regulatory Networks With GINsim 3.0. Front Physiol 2018; 9:646. [PMID: 29971008 PMCID: PMC6018412 DOI: 10.3389/fphys.2018.00646] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/11/2018] [Indexed: 11/13/2022] Open
Abstract
The logical formalism is well adapted to model large cellular networks, in particular when detailed kinetic data are scarce. This tutorial focuses on this well-established qualitative framework. Relying on GINsim (release 3.0), a software implementing this formalism, we guide the reader step by step toward the definition, the analysis and the simulation of a four-node model of the mammalian p53-Mdm2 network.
Collapse
Affiliation(s)
- Aurélien Naldi
- Computational Systems Biology Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), École Normale Supérieure, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Médicale, PSL Université, Paris, France
| | - Céline Hernandez
- Computational Systems Biology Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), École Normale Supérieure, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Médicale, PSL Université, Paris, France
| | - Wassim Abou-Jaoudé
- Computational Systems Biology Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), École Normale Supérieure, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Médicale, PSL Université, Paris, France
| | - Pedro T. Monteiro
- INESC-ID, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | | | - Denis Thieffry
- Computational Systems Biology Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), École Normale Supérieure, Centre National de la Recherche Scientifique, Institut National de la Sante et de la Recherche Médicale, PSL Université, Paris, France
| |
Collapse
|
43
|
Zhang J, Zhang S. The Discovery of Mutated Driver Pathways in Cancer: Models and Algorithms. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:988-998. [PMID: 28113329 DOI: 10.1109/tcbb.2016.2640963] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The pathogenesis of cancer in human is still poorly understood. With the rapid development of high-throughput sequencing technologies, huge volumes of cancer genomics data have been generated. Deciphering that data poses great opportunities and challenges to computational biologists. One of such key challenges is to distinguish driver mutations, genes as well as pathways from passenger ones. Mutual exclusivity of gene mutations (each patient has no more than one mutation in the gene set) has been observed in various cancer types and thus has been used as an important property of a driver gene set or pathway. In this article, we aim to review the recent development of computational models and algorithms for discovering driver pathways or modules in cancer with the focus on mutual exclusivity-based ones.
Collapse
|
44
|
Poret A, Guziolowski C. Therapeutic target discovery using Boolean network attractors: improvements of kali. ROYAL SOCIETY OPEN SCIENCE 2018; 5:171852. [PMID: 29515890 PMCID: PMC5830779 DOI: 10.1098/rsos.171852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/04/2018] [Indexed: 06/10/2023]
Abstract
In a previous article, an algorithm for identifying therapeutic targets in Boolean networks modelling pathological mechanisms was introduced. In the present article, the improvements made on this algorithm, named kali, are described. These improvements are (i) the possibility to work on asynchronous Boolean networks, (ii) a finer assessment of therapeutic targets and (iii) the possibility to use multivalued logic. kali assumes that the attractors of a dynamical system, such as a Boolean network, are associated with the phenotypes of the modelled biological system. Given a logic-based model of pathological mechanisms, kali searches for therapeutic targets able to reduce the reachability of the attractors associated with pathological phenotypes, thus reducing their likeliness. kali is illustrated on an example network and used on a biological case study. The case study is a published logic-based model of bladder tumorigenesis from which kali returns consistent results. However, like any computational tool, kali can predict but cannot replace human expertise: it is a supporting tool for coping with the complexity of biological systems in the field of drug discovery.
Collapse
|
45
|
Lages J, Shepelyansky DL, Zinovyev A. Inferring hidden causal relations between pathway members using reduced Google matrix of directed biological networks. PLoS One 2018; 13:e0190812. [PMID: 29370181 PMCID: PMC5784915 DOI: 10.1371/journal.pone.0190812] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 12/20/2017] [Indexed: 12/18/2022] Open
Abstract
Signaling pathways represent parts of the global biological molecular network which connects them into a seamless whole through complex direct and indirect (hidden) crosstalk whose structure can change during development or in pathological conditions. We suggest a novel methodology, called Googlomics, for the structural analysis of directed biological networks using spectral analysis of their Google matrices, using parallels with quantum scattering theory, developed for nuclear and mesoscopic physics and quantum chaos. We introduce analytical "reduced Google matrix" method for the analysis of biological network structure. The method allows inferring hidden causal relations between the members of a signaling pathway or a functionally related group of genes. We investigate how the structure of hidden causal relations can be reprogrammed as a result of changes in the transcriptional network layer during cancerogenesis. The suggested Googlomics approach rigorously characterizes complex systemic changes in the wiring of large causal biological networks in a computationally efficient way.
Collapse
Affiliation(s)
- José Lages
- Institut UTINAM, Observatoire des Sciences de l'Univers THETA, CNRS, Université de Franche-Comté, 25030 Besançon, France
| | - Dima L Shepelyansky
- Laboratoire de Physique Théorique du CNRS, IRSAMC, Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, F-75005, Paris, France
- Laboratory of advanced methods for high-dimensional data analysis, Lobachevsky University, Nizhni Novgorod, Russia
| |
Collapse
|
46
|
CM-viewer: Visualizing interaction network of co-mutated and mutually exclusively mutated cancer genes. Biosystems 2017; 166:37-42. [PMID: 29278730 DOI: 10.1016/j.biosystems.2017.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 11/19/2017] [Accepted: 12/19/2017] [Indexed: 11/21/2022]
Abstract
Cancer genes usually play a crucial role in regulating cell growth. Normal cells transform into malignant tumors by the acquisition of accumulated genetic mutations that enable them to evade normal growth control. It is therefore important to understand the relationships between mutations during cancer development and progression. Although cancer genes with co-occurring and mutually exclusive mutations have already been studied on different scales, there is no timely updated interaction network available for co-mutated and mutually exclusively mutated cancer genes. Therefore, we firstly downloaded 567 cancer genes from COSMIC (catalogue of somatic mutations in cancer) cancer gene census. Secondly, somatic mutations of 71 cancer genomics projects were downloaded from the ICGC (International Cancer Genome Consortium) data portal. Thirdly, mutated cancer genes and affected donors were extracted from the ICGC data to form a mutation matrix where rows are genes, columns are donors, 1 denotes occurrence, and 0 denotes absence of mutation. Afterwards, co-mutated and mutually exclusively mutated cancer gene pairs were identified using DISCOVER (discrete independence statistic controlling for observations with varying event rates). Finally, CM-viewer was developed to visualize the interaction network of cancer genes with co-occurring and mutually exclusive mutations. It is an online visualization tool as well as a biological database. It promises to understand how gene mutations contribute to tumorigenesis and to identify key biomarkers and drug targets for cancer. CM-viewer is freely available at http://www.zhounan.org/comutgene.
Collapse
|
47
|
Traynard P, Fauré A, Fages F, Thieffry D. Logical model specification aided by model-checking techniques: application to the mammalian cell cycle regulation. Bioinformatics 2017; 32:i772-i780. [PMID: 27587700 DOI: 10.1093/bioinformatics/btw457] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
MOTIVATION Understanding the temporal behaviour of biological regulatory networks requires the integration of molecular information into a formal model. However, the analysis of model dynamics faces a combinatorial explosion as the number of regulatory components and interactions increases. RESULTS We use model-checking techniques to verify sophisticated dynamical properties resulting from the model regulatory structure in the absence of kinetic assumption. We demonstrate the power of this approach by analysing a logical model of the molecular network controlling mammalian cell cycle. This approach enables a systematic analysis of model properties, the delineation of model limitations, and the assessment of various refinements and extensions based on recent experimental observations. The resulting logical model accounts for the main irreversible transitions between cell cycle phases, the sequential activation of cyclins, and the inhibitory role of Skp2, and further emphasizes the multifunctional role for the cell cycle inhibitor Rb. AVAILABILITY AND IMPLEMENTATION The original and revised mammalian cell cycle models are available in the model repository associated with the public modelling software GINsim (http://ginsim.org/node/189). CONTACT thieffry@ens.fr SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Pauline Traynard
- Computational Systems Biology Team, Institut de Biologie de L'Ecole Normale Supérieure (IBENS), CNRS, Inserm, Ecole Normale Supérieure, PSL Research University, Paris, France EPI Lifeware, Inria Inria Saclay Ile-de-France, Palaiseau, France
| | - Adrien Fauré
- Graduate School of Science and Engineering, Yamaguchi University, Yamaguchi, Japan
| | - François Fages
- EPI Lifeware, Inria Inria Saclay Ile-de-France, Palaiseau, France
| | - Denis Thieffry
- Computational Systems Biology Team, Institut de Biologie de L'Ecole Normale Supérieure (IBENS), CNRS, Inserm, Ecole Normale Supérieure, PSL Research University, Paris, France EPI Lifeware, Inria Inria Saclay Ile-de-France, Palaiseau, France
| |
Collapse
|
48
|
Traynard P, Tobalina L, Eduati F, Calzone L, Saez-Rodriguez J. Logic Modeling in Quantitative Systems Pharmacology. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:499-511. [PMID: 28681552 PMCID: PMC5572374 DOI: 10.1002/psp4.12225] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/01/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022]
Abstract
Here we present logic modeling as an approach to understand deregulation of signal transduction in disease and to characterize a drug's mode of action. We discuss how to build a logic model from the literature and experimental data and how to analyze the resulting model to obtain insights of relevance for systems pharmacology. Our workflow uses the free tools OmniPath (network reconstruction from the literature), CellNOpt (model fit to experimental data), MaBoSS (model analysis), and Cytoscape (visualization).
Collapse
Affiliation(s)
- Pauline Traynard
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - Luis Tobalina
- RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, Germany
| | - Federica Eduati
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Laurence Calzone
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - Julio Saez-Rodriguez
- RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, Germany.,European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, UK
| |
Collapse
|
49
|
Zhang J, Zhang S. Discovery of cancer common and specific driver gene sets. Nucleic Acids Res 2017; 45:e86. [PMID: 28168295 PMCID: PMC5449640 DOI: 10.1093/nar/gkx089] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/20/2017] [Accepted: 01/31/2017] [Indexed: 12/31/2022] Open
Abstract
Cancer is known as a disease mainly caused by gene alterations. Discovery of mutated driver pathways or gene sets is becoming an important step to understand molecular mechanisms of carcinogenesis. However, systematically investigating commonalities and specificities of driver gene sets among multiple cancer types is still a great challenge, but this investigation will undoubtedly benefit deciphering cancers and will be helpful for personalized therapy and precision medicine in cancer treatment. In this study, we propose two optimization models to de novo discover common driver gene sets among multiple cancer types (ComMDP) and specific driver gene sets of one certain or multiple cancer types to other cancers (SpeMDP), respectively. We first apply ComMDP and SpeMDP to simulated data to validate their efficiency. Then, we further apply these methods to 12 cancer types from The Cancer Genome Atlas (TCGA) and obtain several biologically meaningful driver pathways. As examples, we construct a common cancer pathway model for BRCA and OV, infer a complex driver pathway model for BRCA carcinogenesis based on common driver gene sets of BRCA with eight cancer types, and investigate specific driver pathways of the liquid cancer lymphoblastic acute myeloid leukemia (LAML) versus other solid cancer types. In these processes more candidate cancer genes are also found.
Collapse
Affiliation(s)
- Junhua Zhang
- National Center for Mathematics and Interdisciplinary Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
| | - Shihua Zhang
- National Center for Mathematics and Interdisciplinary Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematics Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
50
|
Aragon-Ching JB, Trump DL. Targeted therapies in the treatment of urothelial cancers. Urol Oncol 2017; 35:465-472. [PMID: 28366271 DOI: 10.1016/j.urolonc.2017.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 03/04/2017] [Accepted: 03/06/2017] [Indexed: 01/29/2023]
Abstract
Progress has been slow in systemic management of locally advanced and metastatic bladder cancer over the past 20 years. However, the recent approval of immunotherapy with atezolizumab and nivolumab for second-line salvage therapy may usher in an era of more rapid improvement. Systemic treatment is suboptimal and is an area of substantial unmet medical need. The recent findings from The Cancer Genome Atlas project revealed promising pathways that may be amenable to targeted therapies. Promising results with treatment using vascular endothelial growth factor inhibitors such as ramucirumab, sunitinib or bevacizumab, and human epidermal growth factor receptor 2 targeted therapies, epidermal growth factor receptor inhibitors, and fibroblast growth factor receptor inhibitors, are undergoing clinical trials and are discussed later.
Collapse
Affiliation(s)
| | - Donald L Trump
- GU Medical Oncology, Inova Schar Cancer Institute, Fairfax, VA
| |
Collapse
|