1
|
Mizutani T, Boretto M, Lim S, Drost J, González DM, Oka R, Geurts MH, Begthel H, Korving J, van Es JH, van Boxtel R, Clevers H. Recapitulating the adenoma-carcinoma sequence by selection of four spontaneous oncogenic mutations in mismatch-repair-deficient human colon organoids. NATURE CANCER 2024:10.1038/s43018-024-00841-x. [PMID: 39487295 DOI: 10.1038/s43018-024-00841-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 09/23/2024] [Indexed: 11/04/2024]
Abstract
Carcinogenesis results from the sequential acquisition of oncogenic mutations that convert normal cells into invasive, metastasizing cancer cells. Colorectal cancer exemplifies this process through its well-described adenoma-carcinoma sequence, modeled previously using clustered regularly interspaced short palindromic repeats (CRISPR) to induce four consecutive mutations in wild-type human gut organoids. Here, we demonstrate that long-term culture of mismatch-repair-deficient organoids allows the selection of spontaneous oncogenic mutations through the sequential withdrawal of Wnt agonists, epidermal growth factor (EGF) agonists and the bone morphogenetic protein (BMP) antagonist Noggin, while TP53 mutations were selected through the addition of Nutlin-3. Thus, organoids sequentially acquired mutations in AXIN1 and AXIN2 (Wnt pathway), TP53, ACVR2A and BMPR2 (BMP pathway) and NRAS (EGF pathway), gaining complete independence from stem cell niche factors. Quadruple-pathway (Wnt, EGF receptor, p53 and BMP) mutant organoids formed solid tumors upon xenotransplantation. This demonstrates that carcinogenesis can be recapitulated in a DNA repair-mutant background through in vitro selection that targets four consecutive cancer pathways.
Collapse
Affiliation(s)
- Tomohiro Mizutani
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Matteo Boretto
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Sangho Lim
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jarno Drost
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Diego Montiel González
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rurika Oka
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Maarten H Geurts
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Harry Begthel
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Jeroen Korving
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Johan H van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Ruben van Boxtel
- Oncode Institute, Utrecht, The Netherlands
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
- The Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Roche Pharmaceutical Research and Early Development, Basel, Switzerland.
| |
Collapse
|
2
|
Shuaibi A, Chitra U, Raphael BJ. A latent variable model for evaluating mutual exclusivity and co-occurrence between driver mutations in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590995. [PMID: 38712136 PMCID: PMC11071465 DOI: 10.1101/2024.04.24.590995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
A key challenge in cancer genomics is understanding the functional relationships and dependencies between combinations of somatic mutations that drive cancer development. Such driver mutations frequently exhibit patterns of mutual exclusivity or co-occurrence across tumors, and many methods have been developed to identify such dependency patterns from bulk DNA sequencing data of a cohort of patients. However, while mutual exclusivity and co-occurrence are described as properties of driver mutations, existing methods do not explicitly disentangle functional, driver mutations from neutral, passenger mutations. In particular, nearly all existing methods evaluate mutual exclusivity or co-occurrence at the gene level, marking a gene as mutated if any mutation - driver or passenger - is present. Since some genes have a large number of passenger mutations, existing methods either restrict their analyses to a small subset of suspected driver genes - limiting their ability to identify novel dependencies - or make spurious inferences of mutual exclusivity and co-occurrence involving genes with many passenger mutations. We introduce DIALECT, an algorithm to identify dependencies between pairs of driver mutations from somatic mutation counts. We derive a latent variable mixture model for drivers and passengers that combines existing probabilistic models of passenger mutation rates with a latent variable describing the unknown status of a mutation as a driver or passenger. We use an expectation maximization (EM) algorithm to estimate the parameters of our model, including the rates of mutually exclusivity and co-occurrence between drivers. We demonstrate that DIALECT more accurately infers mutual exclusivity and co-occurrence between driver mutations compared to existing methods on both simulated mutation data and somatic mutation data from 5 cancer types in The Cancer Genome Atlas (TCGA).
Collapse
Affiliation(s)
- Ahmed Shuaibi
- Department of Computer Science, Princeton University
- Lewis-Sigler Institute for Integrative Genomics, Princeton University
| | - Uthsav Chitra
- Department of Computer Science, Princeton University
| | | |
Collapse
|
3
|
Antonello A, Bergamin R, Calonaci N, Househam J, Milite S, Williams MJ, Anselmi F, d'Onofrio A, Sundaram V, Sosinsky A, Cross WCH, Caravagna G. Computational validation of clonal and subclonal copy number alterations from bulk tumor sequencing using CNAqc. Genome Biol 2024; 25:38. [PMID: 38297376 PMCID: PMC10832148 DOI: 10.1186/s13059-024-03170-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Copy number alterations (CNAs) are among the most important genetic events in cancer, but their detection from sequencing data is challenging because of unknown sample purity, tumor ploidy, and general intra-tumor heterogeneity. Here, we present CNAqc, an evolution-inspired method to perform the computational validation of clonal and subclonal CNAs detected from bulk DNA sequencing. CNAqc is validated using single-cell data and simulations, is applied to over 4000 TCGA and PCAWG samples, and is incorporated into the validation process for the clinically accredited bioinformatics pipeline at Genomics England. CNAqc is designed to support automated quality control procedures for tumor somatic data validation.
Collapse
Affiliation(s)
- Alice Antonello
- Department of Mathematics, Informatics and Geosciences (MIGe), University of Trieste, Trieste, Italy
| | - Riccardo Bergamin
- Department of Mathematics, Informatics and Geosciences (MIGe), University of Trieste, Trieste, Italy
| | - Nicola Calonaci
- Department of Mathematics, Informatics and Geosciences (MIGe), University of Trieste, Trieste, Italy
| | - Jacob Househam
- Evolution and Cancer Lab, Centre for Genomics and Computational Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Salvatore Milite
- Department of Mathematics, Informatics and Geosciences (MIGe), University of Trieste, Trieste, Italy
- Centre for Computational Biology, Human Technopole, Milan, Italy
| | - Marc J Williams
- Department of Computational Oncology, Memorial Sloan Kettering, New York, USA
| | - Fabio Anselmi
- Department of Mathematics, Informatics and Geosciences (MIGe), University of Trieste, Trieste, Italy
| | - Alberto d'Onofrio
- Department of Mathematics, Informatics and Geosciences (MIGe), University of Trieste, Trieste, Italy
| | | | | | - William C H Cross
- Department of Research Pathology, UCL Cancer Institute, University College London, London, UK
| | - Giulio Caravagna
- Department of Mathematics, Informatics and Geosciences (MIGe), University of Trieste, Trieste, Italy.
- Evolutionary Genomics and Modelling Team, Centre for Evolution and Cancer, Institute of Cancer Research, London, UK.
| |
Collapse
|
4
|
Serebriiskii IG, Pavlov VA, Andrianov GV, Litwin S, Basickes S, Newberg JY, Frampton GM, Meyer JE, Golemis EA. Source, co-occurrence, and prognostic value of PTEN mutations or loss in colorectal cancer. NPJ Genom Med 2023; 8:40. [PMID: 38001126 PMCID: PMC10674024 DOI: 10.1038/s41525-023-00384-7] [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: 08/24/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Somatic PTEN mutations are common and have driver function in some cancer types. However, in colorectal cancers (CRCs), somatic PTEN-inactivating mutations occur at a low frequency (~8-9%), and whether these mutations are actively selected and promote tumor aggressiveness has been controversial. Analysis of genomic data from ~53,000 CRCs indicates that hotspot mutation patterns in PTEN partially reflect DNA-dependent selection pressures, but also suggests a strong selection pressure based on protein function. In microsatellite stable (MSS) tumors, PTEN alterations co-occur with mutations activating BRAF or PI3K, or with TP53 deletions, but not in CRC with microsatellite instability (MSI). Unexpectedly, PTEN deletions are associated with poor survival in MSS CRC, whereas PTEN mutations are associated with improved survival in MSI CRC. These and other data suggest use of PTEN as a prognostic marker is valid in CRC, but such use must consider driver mutation landscape, tumor subtype, and category of PTEN alteration.
Collapse
Affiliation(s)
- Ilya G Serebriiskii
- Program in Cell Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
- Kazan Federal University, 420000, Kazan, Russian Federation.
| | - Valerii A Pavlov
- Program in Cell Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Moscow Institute of Physics and Technology, 141701, Dolgoprudny, Moscow Region, Russian Federation
| | - Grigorii V Andrianov
- Program in Cell Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Samuel Litwin
- Program in Cell Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Stanley Basickes
- Greenfield Manufacturing, 9800 Bustleton Ave, Philadelphia, PA, 19115, USA
| | - Justin Y Newberg
- Foundation Medicine, Inc., 150 Second St., Cambridge, MA, 02141, USA
| | | | - Joshua E Meyer
- Program in Cell Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Program in Cell Signaling and Microenvironment, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
5
|
Fontana D, Crespiatico I, Crippa V, Malighetti F, Villa M, Angaroni F, De Sano L, Aroldi A, Antoniotti M, Caravagna G, Piazza R, Graudenzi A, Mologni L, Ramazzotti D. Evolutionary signatures of human cancers revealed via genomic analysis of over 35,000 patients. Nat Commun 2023; 14:5982. [PMID: 37749078 PMCID: PMC10519956 DOI: 10.1038/s41467-023-41670-3] [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: 03/29/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023] Open
Abstract
Recurring sequences of genomic alterations occurring across patients can highlight repeated evolutionary processes with significant implications for predicting cancer progression. Leveraging the ever-increasing availability of cancer omics data, here we unveil cancer's evolutionary signatures tied to distinct disease outcomes, representing "favored trajectories" of acquisition of driver mutations detected in patients with similar prognosis. We present a framework named ASCETIC (Agony-baSed Cancer EvoluTion InferenCe) to extract such signatures from sequencing experiments generated by different technologies such as bulk and single-cell sequencing data. We apply ASCETIC to (i) single-cell data from 146 myeloid malignancy patients and bulk sequencing from 366 acute myeloid leukemia patients, (ii) multi-region sequencing from 100 early-stage lung cancer patients, (iii) exome/genome data from 10,000+ Pan-Cancer Atlas samples, and (iv) targeted sequencing from 25,000+ MSK-MET metastatic patients, revealing subtype-specific single-nucleotide variant signatures associated with distinct prognostic clusters. Validations on several datasets underscore the robustness and generalizability of the extracted signatures.
Collapse
Affiliation(s)
- Diletta Fontana
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Ilaria Crespiatico
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Valentina Crippa
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Federica Malighetti
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Matteo Villa
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Fabrizio Angaroni
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
- Center of Computational Biology, Human Technopole, Milano, Italy
| | - Luca De Sano
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | - Andrea Aroldi
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Hematology and Clinical Research Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Marco Antoniotti
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre-B4, Milan, Italy
| | - Giulio Caravagna
- Department of Mathematics and Geosciences, University of Trieste, Trieste, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Alex Graudenzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy.
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre-B4, Milan, Italy.
- Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy.
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Daniele Ramazzotti
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| |
Collapse
|
6
|
Li L, Hu Y, Xu Y, Tang S. Mathematical modeling the order of driver gene mutations in colorectal cancer. PLoS Comput Biol 2023; 19:e1011225. [PMID: 37368936 DOI: 10.1371/journal.pcbi.1011225] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Tumor heterogeneity is a large obstacle for cancer study and treatment. Different cancer patients may involve different combinations of gene mutations or the distinct regulatory pathways for inducing the progression of tumor. Investigating the pathways of gene mutations which can cause the formation of tumor can provide a basis for the personalized treatment of cancer. Studies suggested that KRAS, APC and TP53 are the most significant driver genes for colorectal cancer. However, it is still an open issue regarding the detailed mutation order of these genes in the development of colorectal cancer. For this purpose, we analyze the mathematical model considering all orders of mutations in oncogene, KRAS and tumor suppressor genes, APC and TP53, and fit it on data describing the incidence rates of colorectal cancer at different age from the Surveillance Epidemiology and End Results registry in the United States for the year 1973-2013. The specific orders that can induce the development of colorectal cancer are identified by the model fitting. The fitting results indicate that the mutation order with KRAS → APC → TP53, APC → TP53 → KRAS and APC → KRAS → TP53 explain the age-specific risk of colorectal cancer with very well. Furthermore, eleven pathways of gene mutations can be accepted for the mutation order of genes with KRAS → APC → TP53, APC → TP53 → KRAS and APC → KRAS → TP53, and the alternation of APC acts as the initiating or promoting event in the colorectal cancer. The estimated mutation rates of cells in the different pathways demonstrate that genetic instability must exist in colorectal cancer with alterations of genes, KRAS, APC and TP53.
Collapse
Affiliation(s)
- Lingling Li
- School of Mathematics and Statistics, Shaanxi Normal University, Xi'an, China
- School of Science, Xi'an Polytechnic University, Xi'an, China
| | - Yulu Hu
- School of Science, Xi'an Polytechnic University, Xi'an, China
| | - Yunshan Xu
- Mathematics Department, Faculty of Science and Technology, University of Macau, Taipa, Macau, China
| | - Sanyi Tang
- School of Mathematics and Statistics, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
7
|
Proietto M, Crippa M, Damiani C, Pasquale V, Sacco E, Vanoni M, Gilardi M. Tumor heterogeneity: preclinical models, emerging technologies, and future applications. Front Oncol 2023; 13:1164535. [PMID: 37188201 PMCID: PMC10175698 DOI: 10.3389/fonc.2023.1164535] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Heterogeneity describes the differences among cancer cells within and between tumors. It refers to cancer cells describing variations in morphology, transcriptional profiles, metabolism, and metastatic potential. More recently, the field has included the characterization of the tumor immune microenvironment and the depiction of the dynamics underlying the cellular interactions promoting the tumor ecosystem evolution. Heterogeneity has been found in most tumors representing one of the most challenging behaviors in cancer ecosystems. As one of the critical factors impairing the long-term efficacy of solid tumor therapy, heterogeneity leads to tumor resistance, more aggressive metastasizing, and recurrence. We review the role of the main models and the emerging single-cell and spatial genomic technologies in our understanding of tumor heterogeneity, its contribution to lethal cancer outcomes, and the physiological challenges to consider in designing cancer therapies. We highlight how tumor cells dynamically evolve because of the interactions within the tumor immune microenvironment and how to leverage this to unleash immune recognition through immunotherapy. A multidisciplinary approach grounded in novel bioinformatic and computational tools will allow reaching the integrated, multilayered knowledge of tumor heterogeneity required to implement personalized, more efficient therapies urgently required for cancer patients.
Collapse
Affiliation(s)
- Marco Proietto
- Next Generation Sequencing Core, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Martina Crippa
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Imaging Center, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Chiara Damiani
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Valentina Pasquale
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Elena Sacco
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Marco Vanoni
- Infrastructure Systems Biology Europe /Centre of Systems Biology (ISBE/SYSBIO) Centre of Systems Biology, Milan, Italy
- Department of Biotechnology and Biosciences, School of Sciences, University of Milano-Bicocca, Milan, Italy
| | - Mara Gilardi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Salk Cancer Center, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
8
|
Li H, Sun Z, Xiao R, Qi Q, Li X, Huang H, Wang X, Zhou J, Wang Z, Liu K, Yin P, Yang F, Wang J. Stepwise evolutionary genomics of early-stage lung adenocarcinoma manifesting as pure, heterogeneous and part-solid ground-glass nodules. Br J Cancer 2022; 127:747-756. [PMID: 35618790 PMCID: PMC9381762 DOI: 10.1038/s41416-022-01821-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/25/2022] [Accepted: 04/04/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This study was designed to unravel the genomic landscape and evolution of early-stage subsolid lung adenocarcinomas (SSN-LUADs) manifesting as pure ground-glass nodules (pGGNs), heterogeneous ground-glass nodules (HGGNs) and part-solid nodules (PSNs). METHODS Samples subjected to either broad-panel next-generation sequencing (NGS) or whole-exome sequencing (WES) were included. Clinicopathologic and genomic features were compared among pGGN, HGGN and PSN, while tumour evolutionary trajectories and mutational signatures were evaluated in the entire cohort. RESULTS In total, 247 SSN-LUAD samples subjected to broad-panel NGS and 125 to WES were identified. Compared with PSNs, HGGNs had significantly lower tumour mutation count (P < 0.001), genomic alteration count (P < 0.001), and intra-tumour heterogeneity (P = 0.005). Statistically significant upward trends were observed in alterations involving driver mutations and oncogenic pathways from pGGNs to HGGNs to PSNs. EGFR mutation was proved to be a key early event in the progression of SSN-LUADs, with subsequently two evolutionary trajectories involving either RBM10 or TP53 mutation in the cancer-evolution models. CONCLUSIONS This study provided evidence for unravelling the previously unknown genomic underpinnings associated with SSN-LUAD evolution from pGGN to HGGN to PSN, proving that HGGN was an intermediate SSN form between pGGN and PSN genetically.
Collapse
Affiliation(s)
- Hao Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Zewen Sun
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Rongxin Xiao
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Qingyi Qi
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Xiao Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China.
| | - Haiyan Huang
- Berry Oncology Corporation, No. 4 Science Park Road, Changping District, Beijing, China
| | - Xuan Wang
- Department of Cardiac Surgery, Peking University People's Hospital, Beijing, China
| | - Jian Zhou
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Zhenfan Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Ke Liu
- Berry Oncology Corporation, No. 4 Science Park Road, Changping District, Beijing, China
| | - Ping Yin
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
| | - Jun Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
9
|
Angaroni F, Guidi A, Ascolani G, d'Onofrio A, Antoniotti M, Graudenzi A. J-SPACE: a Julia package for the simulation of spatial models of cancer evolution and of sequencing experiments. BMC Bioinformatics 2022; 23:269. [PMID: 35804300 PMCID: PMC9270769 DOI: 10.1186/s12859-022-04779-8] [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: 02/23/2022] [Accepted: 06/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background The combined effects of biological variability and measurement-related errors on cancer sequencing data remain largely unexplored. However, the spatio-temporal simulation of multi-cellular systems provides a powerful instrument to address this issue. In particular, efficient algorithmic frameworks are needed to overcome the harsh trade-off between scalability and expressivity, so to allow one to simulate both realistic cancer evolution scenarios and the related sequencing experiments, which can then be used to benchmark downstream bioinformatics methods. Result We introduce a Julia package for SPAtial Cancer Evolution (J-SPACE), which allows one to model and simulate a broad set of experimental scenarios, phenomenological rules and sequencing settings.Specifically, J-SPACE simulates the spatial dynamics of cells as a continuous-time multi-type birth-death stochastic process on a arbitrary graph, employing different rules of interaction and an optimised Gillespie algorithm. The evolutionary dynamics of genomic alterations (single-nucleotide variants and indels) is simulated either under the Infinite Sites Assumption or several different substitution models, including one based on mutational signatures. After mimicking the spatial sampling of tumour cells, J-SPACE returns the related phylogenetic model, and allows one to generate synthetic reads from several Next-Generation Sequencing (NGS) platforms, via the ART read simulator. The results are finally returned in standard FASTA, FASTQ, SAM, ALN and Newick file formats. Conclusion J-SPACE is designed to efficiently simulate the heterogeneous behaviour of a large number of cancer cells and produces a rich set of outputs. Our framework is useful to investigate the emergent spatial dynamics of cancer subpopulations, as well as to assess the impact of incomplete sampling and of experiment-specific errors. Importantly, the output of J-SPACE is designed to allow the performance assessment of downstream bioinformatics pipelines processing NGS data. J-SPACE is freely available at: https://github.com/BIMIB-DISCo/J-Space.jl.
Collapse
Affiliation(s)
- Fabrizio Angaroni
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy.
| | - Alessandro Guidi
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy
| | - Gianluca Ascolani
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy
| | - Alberto d'Onofrio
- Department of Mathematics and Geosciences, Univ. of Trieste, Trieste, Italy
| | - Marco Antoniotti
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre (B4), Milan, Italy
| | - Alex Graudenzi
- Dept. of Informatics, Systems and Communication, Univ. of Milan-Bicocca, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre (B4), Milan, Italy.,Inst. of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Segrate, Italy
| |
Collapse
|
10
|
Yan J, Hu Z, Li ZW, Sun S, Guo WF. Network Control Models With Personalized Genomics Data for Understanding Tumor Heterogeneity in Cancer. Front Oncol 2022; 12:891676. [PMID: 35712516 PMCID: PMC9195174 DOI: 10.3389/fonc.2022.891676] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022] Open
Abstract
Due to rapid development of high-throughput sequencing and biotechnology, it has brought new opportunities and challenges in developing efficient computational methods for exploring personalized genomics data of cancer patients. Because of the high-dimension and small sample size characteristics of these personalized genomics data, it is difficult for excavating effective information by using traditional statistical methods. In the past few years, network control methods have been proposed to solve networked system with high-dimension and small sample size. Researchers have made progress in the design and optimization of network control principles. However, there are few studies comprehensively surveying network control methods to analyze the biomolecular network data of individual patients. To address this problem, here we comprehensively surveyed complex network control methods on personalized omics data for understanding tumor heterogeneity in precision medicine of individual patients with cancer.
Collapse
Affiliation(s)
- Jipeng Yan
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Zhuo Hu
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
| | - Zong-Wei Li
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Wei-Feng Guo, ; Shiren Sun,
| | - Wei-Feng Guo
- School of Electrical Engineering, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Wei-Feng Guo, ; Shiren Sun,
| |
Collapse
|
11
|
Calabretta E, Guidetti A, Ricci F, Di Trani M, Monfrini C, Magagnoli M, Bramanti S, Maspero D, Morello L, Merli M, Di Rocco A, Graudenzi A, Derenzini E, Antoniotti M, Rossi D, Corradini P, Santoro A, Carlo-Stella C. Chemotherapy after PD-1 inhibitors in relapsed/refractory Hodgkin lymphoma: Outcomes and clonal evolution dynamics. Br J Haematol 2022; 198:82-92. [PMID: 35468225 PMCID: PMC9321573 DOI: 10.1111/bjh.18183] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 11/27/2022]
Abstract
Checkpoint inhibitors (CPIs) are routinely employed in relapsed/refractory classical Hodgkin lymphoma. Nonetheless, persistent long-term responses are uncommon, and one-third of patients are refractory. Several reports have suggested that treatment with CPIs may re-sensitize patients to chemotherapy, however there is no consensus on the optimal chemotherapy regimen and subsequent consolidation strategy. In this retrospective study we analysed the response to rechallenge with chemotherapy after CPI failure. Furthermore, we exploratively characterized the clonal evolution profile of a small sample of patients (n = 5) by employing the CALDER approach. Among the 28 patients included in the study, 17 (71%) were primary refractory and 26 (92%) were refractory to the last chemotherapy prior to CPIs. Following rechallenge with chemotherapy, response was recorded in 23 (82%) patients experiencing complete remission and 3 (11%) patients experiencing partial remission. The tumour evolution of the patients inferred by CALDER seemingly occurred prior to the first cycle of therapy and was characterized either by linear or branching evolution patterns. Twenty-five patients proceeded to allogeneic stem cell transplantation. At a median follow-up of 21 months, median PFS and OS were not reached. In conclusion, patients who fail CPIs can be effectively rescued by salvage chemotherapy and bridged to allo-SCT/auto-SCT.
Collapse
Affiliation(s)
- Eleonora Calabretta
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Anna Guidetti
- Division of Hematology and Bone Marrow Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,University of Milano, Milan, Italy
| | - Francesca Ricci
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Martina Di Trani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Chiara Monfrini
- Division of Hematology and Bone Marrow Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Massimo Magagnoli
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Stefania Bramanti
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Davide Maspero
- Dipartimento di Informatica, Sistemistica e Comunicazione, Università degli Studi di Milano-Bicocca, Milan, Italy.,Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
| | - Lucia Morello
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Michele Merli
- Department of Hematology, University Hospital "Ospedale di Circolo e Fondazione Macchi - ASST Sette Laghi", University of Insubria, Varese, Italy
| | - Alice Di Rocco
- Department of Translational and Precision Medicine, La Sapienza University, Rome, Italy
| | - Alex Graudenzi
- Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre (B4), Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Enrico Derenzini
- IEO European Institute of Oncology IRCCS, Milan, Italy.,Department of Health Sciences, University of Milan, Italy
| | - Marco Antoniotti
- Dipartimento di Informatica, Sistemistica e Comunicazione, Università degli Studi di Milano-Bicocca, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre (B4), Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Davide Rossi
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.,Institute of Oncology Research, Bellinzona, Switzerland.,Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Paolo Corradini
- Division of Hematology and Bone Marrow Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,University of Milano, Milan, Italy
| | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
12
|
Milite S, Bergamin R, Patruno L, Calonaci N, Caravagna G. A Bayesian method to cluster single-cell RNA sequencing data using Copy Number Alterations. Bioinformatics 2022; 38:2512-2518. [PMID: 35298589 DOI: 10.1093/bioinformatics/btac143] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 01/31/2022] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Cancers are composed by several heterogeneous subpopulations, each one harbouring different genetic and epigenetic somatic alterations that contribute to disease onset and therapy response. In recent years, copy number alterations leading to tumour aneuploidy have been identified as potential key drivers of such populations, but the definition of the precise makeup of cancer subclones from sequencing assays remains challenging. In the end, little is known about the mapping between complex copy number alterations and their effect on cancer phenotypes. RESULTS We introduce CONGAS, a Bayesian probabilistic method to phase bulk DNA and single-cell RNA measurements from independent assays. CONGAS jointly identifies clusters of single cells with subclonal copy number alterations, and differences in RNA expression. The model builds statistical priors leveraging bulk DNA sequencing data, does not require a normal reference and scales fast thanks to a GPU backend and variational inference. We test CONGAS on both simulated and real data, and find that it can determine the tumour subclonal composition at the single-cell level together with clone-specific RNA phenotypes in tumour data generated from both 10x and Smart-Seq assays. AVAILABILITY CONGAS is available as 2 packages: CONGAS (https://github.com/caravagnalab/congas), which implements the model in Python, and RCONGAS (https://caravagnalab.github.io/rcongas/), which provides R functions to process inputs, outputs, and run CONGAS fits. The analysis of real data and scripts to generate figures of this paper are available via RCONGAS; code associated to simulations is available at https://github.com/caravagnalab/rcongas_test. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Salvatore Milite
- Department of Mathematics and Geosciences, University of Trieste, Italy
| | - Riccardo Bergamin
- Department of Mathematics and Geosciences, University of Trieste, Italy
| | - Lucrezia Patruno
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Italy
| | - Nicola Calonaci
- Department of Mathematics and Geosciences, University of Trieste, Italy
| | - Giulio Caravagna
- Department of Mathematics and Geosciences, University of Trieste, Italy
| |
Collapse
|
13
|
Li H, Sun Z, Li Y, Qi Q, Huang H, Wang X, Zhou J, Liu K, Yin P, Wang Z, Li X, Yang F. Disparate Genomic Characteristics of Patients with Early-Stage Lung Adenocarcinoma Manifesting as Radiological Subsolid or Solid Lesions. Lung Cancer 2022; 166:178-188. [DOI: 10.1016/j.lungcan.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/21/2022] [Accepted: 02/22/2022] [Indexed: 11/27/2022]
|
14
|
Early Breast Cancer Evolution by Autosomal Broad Copy Number Alterations. Int J Genomics 2022; 2022:9332922. [PMID: 35252434 PMCID: PMC8896957 DOI: 10.1155/2022/9332922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
The availability of comprehensive genomic datasets across patient populations enables the application of novel methods for reconstructing tumor evolution within individual patients. To this end, we propose studying autosomal broad copy number alterations (CNAs) as a framework to better understand early tumor evolution. We compared the broad CNAs and somatic mutations of patients with 1 to 10 autosomal broad CNAs against the full set of patients, using data from The Cancer Genome Atlas breast cancer project. We reveal here that the frequency of a chromosome arm obtaining a broad CNA and a genome acquiring somatic mutations changes as autosomal broad CNAs accumulate. Therefore, we propose that the number of autosomal broad CNAs is an important characteristic of breast tumors that needs to be taken into consideration when studying breast tumors. To investigate this idea more in-depth, we next studied the frequency that specific chromosome arms acquire broad CNAs in patients with 1 to 10 broad CNAs. With this process, we identified the broad CNAs that exhibit the fastest rates of accumulation across all patients. This finding suggests a likely order of occurrence of these alterations in patients, which is apparent when we consider a subset of patients with few broad CNAs. Here, we lay the foundation for future studies to build upon our findings and use autosomal broad CNAs as a method to monitor breast tumor progression in vivo to further our understanding of how early tumor evolution unfolds.
Collapse
|
15
|
Angaroni F, Chen K, Damiani C, Caravagna G, Graudenzi A, Ramazzotti D. PMCE: efficient inference of expressive models of cancer evolution with high prognostic power. Bioinformatics 2022; 38:754-762. [PMID: 34647978 DOI: 10.1093/bioinformatics/btab717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Driver (epi)genomic alterations underlie the positive selection of cancer subpopulations, which promotes drug resistance and relapse. Even though substantial heterogeneity is witnessed in most cancer types, mutation accumulation patterns can be regularly found and can be exploited to reconstruct predictive models of cancer evolution. Yet, available methods can not infer logical formulas connecting events to represent alternative evolutionary routes or convergent evolution. RESULTS We introduce PMCE, an expressive framework that leverages mutational profiles from cross-sectional sequencing data to infer probabilistic graphical models of cancer evolution including arbitrary logical formulas, and which outperforms the state-of-the-art in terms of accuracy and robustness to noise, on simulations. The application of PMCE to 7866 samples from the TCGA database allows us to identify a highly significant correlation between the predicted evolutionary paths and the overall survival in 7 tumor types, proving that our approach can effectively stratify cancer patients in reliable risk groups. AVAILABILITY AND IMPLEMENTATION PMCE is freely available at https://github.com/BIMIB-DISCo/PMCE, in addition to the code to replicate all the analyses presented in the manuscript. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Fabrizio Angaroni
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan 20125, Italy
| | - Kevin Chen
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Chiara Damiani
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan 20126, Italy.,Sysbio Centre for Systems Biology, Milan 20100, Italy
| | - Giulio Caravagna
- Department of Mathematics and Geosciences, University of Trieste, Trieste 34128, Italy
| | - Alex Graudenzi
- Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan 20054, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre (B4), Milan 20100, Italy
| | - Daniele Ramazzotti
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA.,Department of Pathology, Stanford University, Stanford, CA 94305, USA.,Department of Medicine and Surgery, University of Milan-Bicocca, Monza 20900, Italy
| |
Collapse
|
16
|
Diaz-Colunga J, Diaz-Uriarte R. Conditional prediction of consecutive tumor evolution using cancer progression models: What genotype comes next? PLoS Comput Biol 2021; 17:e1009055. [PMID: 34932572 PMCID: PMC8730404 DOI: 10.1371/journal.pcbi.1009055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 01/05/2022] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Accurate prediction of tumor progression is key for adaptive therapy and precision medicine. Cancer progression models (CPMs) can be used to infer dependencies in mutation accumulation from cross-sectional data and provide predictions of tumor progression paths. However, their performance when predicting complete evolutionary trajectories is limited by violations of assumptions and the size of available data sets. Instead of predicting full tumor progression paths, here we focus on short-term predictions, more relevant for diagnostic and therapeutic purposes. We examine whether five distinct CPMs can be used to answer the question "Given that a genotype with n mutations has been observed, what genotype with n + 1 mutations is next in the path of tumor progression?" or, shortly, "What genotype comes next?". Using simulated data we find that under specific combinations of genotype and fitness landscape characteristics CPMs can provide predictions of short-term evolution that closely match the true probabilities, and that some genotype characteristics can be much more relevant than global features. Application of these methods to 25 cancer data sets shows that their use is hampered by a lack of information needed to make principled decisions about method choice. Fruitful use of these methods for short-term predictions requires adapting method's use to local genotype characteristics and obtaining reliable indicators of performance; it will also be necessary to clarify the interpretation of the method's results when key assumptions do not hold.
Collapse
Affiliation(s)
- Juan Diaz-Colunga
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (UAM-CSIC), Madrid, Spain
- Department of Ecology & Evolutionary Biology and Microbial Sciences Institute, Yale University, New Haven, Connecticut, United States of America
| | - Ramon Diaz-Uriarte
- Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (UAM-CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
17
|
Nobile MS, Cazzaniga P, Ramazzotti D. Investigating the performance of multi-objective optimization when learning Bayesian Networks. Neurocomputing 2021. [DOI: 10.1016/j.neucom.2021.07.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
18
|
Guo WF, Zhang SW, Zeng T, Akutsu T, Chen L. Network control principles for identifying personalized driver genes in cancer. Brief Bioinform 2021; 21:1641-1662. [PMID: 31711128 DOI: 10.1093/bib/bbz089] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 02/02/2023] Open
Abstract
To understand tumor heterogeneity in cancer, personalized driver genes (PDGs) need to be identified for unraveling the genotype-phenotype associations corresponding to particular patients. However, most of the existing driver-focus methods mainly pay attention on the cohort information rather than on individual information. Recent developing computational approaches based on network control principles are opening a new way to discover driver genes in cancer, particularly at an individual level. To provide comprehensive perspectives of network control methods on this timely topic, we first considered the cancer progression as a network control problem, in which the expected PDGs are altered genes by oncogene activation signals that can change the individual molecular network from one health state to the other disease state. Then, we reviewed the network reconstruction methods on single samples and introduced novel network control methods on single-sample networks to identify PDGs in cancer. Particularly, we gave a performance assessment of the network structure control-based PDGs identification methods on multiple cancer datasets from TCGA, for which the data and evaluation package also are publicly available. Finally, we discussed future directions for the application of network control methods to identify PDGs in cancer and diverse biological processes.
Collapse
Affiliation(s)
- Wei-Feng Guo
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Shao-Wu Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji, 611-0011, Japan
| | - Luonan Chen
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi'an 710072, China.,Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, 201210 Shanghai, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
19
|
|
20
|
Li L, Shao M, He X, Ren S, Tian T. Risk of lung cancer due to external environmental factor and epidemiological data analysis. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:6079-6094. [PMID: 34517524 DOI: 10.3934/mbe.2021304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Lung cancer is a cancer with the fastest growth in the incidence and mortality all over the world, which is an extremely serious threat to human's life and health. Evidences reveal that external environmental factors are the key drivers of lung cancer, such as smoking, radiation exposure and so on. Therefore, it is urgent to explain the mechanism of lung cancer risk due to external environmental factors experimentally and theoretically. However, it is still an open issue regarding how external environment factors affect lung cancer risk. In this paper, we summarize the main mathematical models involved the gene mutations for cancers, and review the application of the models to analyze the mechanism of lung cancer and the risk of lung cancer due to external environmental exposure. In addition, we apply the model described and the epidemiological data to analyze the influence of external environmental factors on lung cancer risk. The result indicates that radiation can cause significantly an increase in the mutation rate of cells, in particular the mutation in stability gene that leads to genomic instability. These studies not only can offer insights into the relationship between external environmental factors and human lung cancer risk, but also can provide theoretical guidance for the prevention and control of lung cancer.
Collapse
Affiliation(s)
- Lingling Li
- School of Science, Xi'an Polytechnic University, Xi'an 710048, China
| | - Mengyao Shao
- School of Science, Xi'an Polytechnic University, Xi'an 710048, China
| | - Xingshi He
- School of Science, Xi'an Polytechnic University, Xi'an 710048, China
| | - Shanjing Ren
- School of Mathematics and Big Data, GuiZhou Education University, Guiyang 550018, China
| | - Tianhai Tian
- School of Mathematical Science, Monash University, Melbourne Vic 3800, Australia
| |
Collapse
|
21
|
Nicol PB, Coombes KR, Deaver C, Chkrebtii O, Paul S, Toland AE, Asiaee A. Oncogenetic network estimation with disjunctive Bayesian networks. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021. [DOI: 10.1002/cso2.1027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
| | - Kevin R. Coombes
- Department of Biomedical Informatics Ohio State University Columbus Ohio
| | - Courtney Deaver
- Natural Sciences Division Pepperdine University Malibu California
| | | | - Subhadeep Paul
- Department of Statistics Ohio State University Columbus Ohio
| | - Amanda E. Toland
- Department of Cancer Biology and Genetics and Department of Internal Medicine Division of Human Genetics, Comprehensive Cancer Center Ohio State University Columbus Ohio
| | - Amir Asiaee
- Mathematical Biosciences Institute Ohio State University Columbus Ohio
| |
Collapse
|
22
|
Manrubia S, Cuesta JA, Aguirre J, Ahnert SE, Altenberg L, Cano AV, Catalán P, Diaz-Uriarte R, Elena SF, García-Martín JA, Hogeweg P, Khatri BS, Krug J, Louis AA, Martin NS, Payne JL, Tarnowski MJ, Weiß M. From genotypes to organisms: State-of-the-art and perspectives of a cornerstone in evolutionary dynamics. Phys Life Rev 2021; 38:55-106. [PMID: 34088608 DOI: 10.1016/j.plrev.2021.03.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/01/2021] [Indexed: 12/21/2022]
Abstract
Understanding how genotypes map onto phenotypes, fitness, and eventually organisms is arguably the next major missing piece in a fully predictive theory of evolution. We refer to this generally as the problem of the genotype-phenotype map. Though we are still far from achieving a complete picture of these relationships, our current understanding of simpler questions, such as the structure induced in the space of genotypes by sequences mapped to molecular structures, has revealed important facts that deeply affect the dynamical description of evolutionary processes. Empirical evidence supporting the fundamental relevance of features such as phenotypic bias is mounting as well, while the synthesis of conceptual and experimental progress leads to questioning current assumptions on the nature of evolutionary dynamics-cancer progression models or synthetic biology approaches being notable examples. This work delves with a critical and constructive attitude into our current knowledge of how genotypes map onto molecular phenotypes and organismal functions, and discusses theoretical and empirical avenues to broaden and improve this comprehension. As a final goal, this community should aim at deriving an updated picture of evolutionary processes soundly relying on the structural properties of genotype spaces, as revealed by modern techniques of molecular and functional analysis.
Collapse
Affiliation(s)
- Susanna Manrubia
- Department of Systems Biology, Centro Nacional de Biotecnología (CSIC), Madrid, Spain; Grupo Interdisciplinar de Sistemas Complejos (GISC), Madrid, Spain.
| | - José A Cuesta
- Grupo Interdisciplinar de Sistemas Complejos (GISC), Madrid, Spain; Departamento de Matemáticas, Universidad Carlos III de Madrid, Leganés, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BiFi), Universidad de Zaragoza, Spain; UC3M-Santander Big Data Institute (IBiDat), Getafe, Madrid, Spain
| | - Jacobo Aguirre
- Grupo Interdisciplinar de Sistemas Complejos (GISC), Madrid, Spain; Centro de Astrobiología, CSIC-INTA, ctra. de Ajalvir km 4, 28850 Torrejón de Ardoz, Madrid, Spain
| | - Sebastian E Ahnert
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, UK; The Alan Turing Institute, British Library, 96 Euston Road, London NW1 2DB, UK
| | | | - Alejandro V Cano
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Pablo Catalán
- Grupo Interdisciplinar de Sistemas Complejos (GISC), Madrid, Spain; Departamento de Matemáticas, Universidad Carlos III de Madrid, Leganés, Spain
| | - Ramon Diaz-Uriarte
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain; Instituto de Investigaciones Biomédicas "Alberto Sols" (UAM-CSIC), Madrid, Spain
| | - Santiago F Elena
- Instituto de Biología Integrativa de Sistemas, I(2)SysBio (CSIC-UV), València, Spain; The Santa Fe Institute, Santa Fe, NM, USA
| | | | - Paulien Hogeweg
- Theoretical Biology and Bioinformatics Group, Utrecht University, the Netherlands
| | - Bhavin S Khatri
- The Francis Crick Institute, London, UK; Department of Life Sciences, Imperial College London, London, UK
| | - Joachim Krug
- Institute for Biological Physics, University of Cologne, Köln, Germany
| | - Ard A Louis
- Rudolf Peierls Centre for Theoretical Physics, University of Oxford, Oxford, UK
| | - Nora S Martin
- Theory of Condensed Matter Group, Cavendish Laboratory, University of Cambridge, Cambridge, UK; Sainsbury Laboratory, University of Cambridge, Cambridge, UK
| | - Joshua L Payne
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Marcel Weiß
- Theory of Condensed Matter Group, Cavendish Laboratory, University of Cambridge, Cambridge, UK; Sainsbury Laboratory, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
Ramazzotti D, Angaroni F, Maspero D, Gambacorti-Passerini C, Antoniotti M, Graudenzi A, Piazza R. VERSO: A comprehensive framework for the inference of robust phylogenies and the quantification of intra-host genomic diversity of viral samples. PATTERNS (NEW YORK, N.Y.) 2021; 2:100212. [PMID: 33728416 PMCID: PMC7953447 DOI: 10.1016/j.patter.2021.100212] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/30/2020] [Accepted: 01/22/2021] [Indexed: 12/22/2022]
Abstract
We introduce VERSO, a two-step framework for the characterization of viral evolution from sequencing data of viral genomes, which is an improvement on phylogenomic approaches for consensus sequences. VERSO exploits an efficient algorithmic strategy to return robust phylogenies from clonal variant profiles, also in conditions of sampling limitations. It then leverages variant frequency patterns to characterize the intra-host genomic diversity of samples, revealing undetected infection chains and pinpointing variants likely involved in homoplasies. On simulations, VERSO outperforms state-of-the-art tools for phylogenetic inference. Notably, the application to 6,726 amplicon and RNA sequencing samples refines the estimation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolution, while co-occurrence patterns of minor variants unveil undetected infection paths, which are validated with contact tracing data. Finally, the analysis of SARS-CoV-2 mutational landscape uncovers a temporal increase of overall genomic diversity and highlights variants transiting from minor to clonal state and homoplastic variants, some of which fall on the spike gene. Available at: https://github.com/BIMIB-DISCo/VERSO.
Collapse
Affiliation(s)
- Daniele Ramazzotti
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - Fabrizio Angaroni
- Department of Informatics, Systems and Communication, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Davide Maspero
- Department of Informatics, Systems and Communication, Università degli Studi di Milano-Bicocca, Milan, Italy
- Inst. of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
| | | | - Marco Antoniotti
- Department of Informatics, Systems and Communication, Università degli Studi di Milano-Bicocca, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre – B4, Milan, Italy
| | - Alex Graudenzi
- Inst. of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre – B4, Milan, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| |
Collapse
|
24
|
Machicao J, Craighero F, Maspero D, Angaroni F, Damiani C, Graudenzi A, Antoniotti M, Bruno OM. On the Use of Topological Features of Metabolic Networks for the Classification of Cancer Samples. Curr Genomics 2021; 22:88-97. [PMID: 34220296 PMCID: PMC8188584 DOI: 10.2174/1389202922666210301084151] [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: 07/07/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The increasing availability of omics data collected from patients affected by severe pathologies, such as cancer, is fostering the development of data science methods for their analysis. INTRODUCTION The combination of data integration and machine learning approaches can provide new powerful instruments to tackle the complexity of cancer development and deliver effective diagnostic and prognostic strategies. METHODS We explore the possibility of exploiting the topological properties of sample-specific metabolic networks as features in a supervised classification task. Such networks are obtained by projecting transcriptomic data from RNA-seq experiments on genome-wide metabolic models to define weighted networks modeling the overall metabolic activity of a given sample. RESULTS We show the classification results on a labeled breast cancer dataset from the TCGA database, including 210 samples (cancer vs. normal). In particular, we investigate how the performance is affected by a threshold-based pruning of the networks by comparing Artificial Neural Networks, Support Vector Machines and Random Forests. Interestingly, the best classification performance is achieved within a small threshold range for all methods, suggesting that it might represent an effective choice to recover useful information while filtering out noise from data. Overall, the best accuracy is achieved with SVMs, which exhibit performances similar to those obtained when gene expression profiles are used as features. CONCLUSION These findings demonstrate that the topological properties of sample-specific metabolic networks are effective in classifying cancer and normal samples, suggesting that useful information can be extracted from a relatively limited number of features.
Collapse
Affiliation(s)
- Jeaneth Machicao
- Address correspondence to these authors at the São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil; Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy E-mails: , ,
| | | | | | | | | | - Alex Graudenzi
- Address correspondence to these authors at the São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil; Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy E-mails: , ,
| | | | - Odemir M. Bruno
- Address correspondence to these authors at the São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil; Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy E-mails: , ,
| |
Collapse
|
25
|
Measuring evolutionary cancer dynamics from genome sequencing, one patient at a time. Stat Appl Genet Mol Biol 2020. [DOI: 10.1515/sagmb-2020-0075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AbstractCancers progress through the accumulation of somatic mutations which accrue during tumour evolution, allowing some cells to proliferate in an uncontrolled fashion. This growth process is intimately related to latent evolutionary forces moulding the genetic and epigenetic composition of tumour subpopulations. Understanding cancer requires therefore the understanding of these selective pressures. The adoption of widespread next-generation sequencing technologies opens up for the possibility of measuring molecular profiles of cancers at multiple resolutions, across one or multiple patients. In this review we discuss how cancer genome sequencing data from a single tumour can be used to understand these evolutionary forces, overviewing mathematical models and inferential methods adopted in field of Cancer Evolution.
Collapse
|
26
|
Kondylakis H, Axenie C, Kiran Bastola D, Katehakis DG, Kouroubali A, Kurz D, Larburu N, Macía I, Maguire R, Maramis C, Marias K, Morrow P, Muro N, Núñez-Benjumea FJ, Rampun A, Rivera-Romero O, Scotney B, Signorelli G, Wang H, Tsiknakis M, Zwiggelaar R. Status and Recommendations of Technological and Data-Driven Innovations in Cancer Care: Focus Group Study. J Med Internet Res 2020; 22:e22034. [PMID: 33320099 PMCID: PMC7772066 DOI: 10.2196/22034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/02/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The status of the data-driven management of cancer care as well as the challenges, opportunities, and recommendations aimed at accelerating the rate of progress in this field are topics of great interest. Two international workshops, one conducted in June 2019 in Cordoba, Spain, and one in October 2019 in Athens, Greece, were organized by four Horizon 2020 (H2020) European Union (EU)-funded projects: BOUNCE, CATCH ITN, DESIREE, and MyPal. The issues covered included patient engagement, knowledge and data-driven decision support systems, patient journey, rehabilitation, personalized diagnosis, trust, assessment of guidelines, and interoperability of information and communication technology (ICT) platforms. A series of recommendations was provided as the complex landscape of data-driven technical innovation in cancer care was portrayed. OBJECTIVE This study aims to provide information on the current state of the art of technology and data-driven innovations for the management of cancer care through the work of four EU H2020-funded projects. METHODS Two international workshops on ICT in the management of cancer care were held, and several topics were identified through discussion among the participants. A focus group was formulated after the second workshop, in which the status of technological and data-driven cancer management as well as the challenges, opportunities, and recommendations in this area were collected and analyzed. RESULTS Technical and data-driven innovations provide promising tools for the management of cancer care. However, several challenges must be successfully addressed, such as patient engagement, interoperability of ICT-based systems, knowledge management, and trust. This paper analyzes these challenges, which can be opportunities for further research and practical implementation and can provide practical recommendations for future work. CONCLUSIONS Technology and data-driven innovations are becoming an integral part of cancer care management. In this process, specific challenges need to be addressed, such as increasing trust and engaging the whole stakeholder ecosystem, to fully benefit from these innovations.
Collapse
Affiliation(s)
| | - Cristian Axenie
- Audi Konfuzius-Institut Ingolstadt Lab, Technische Hochschule Ingolstadt, Ingolstadt, Germany
| | - Dhundy Kiran Bastola
- School of Interdisciplinary Informatics, University of Nebraska, Omaha, NE, United States
| | | | | | - Daria Kurz
- Interdisziplinäres Brustzentrum, Helios Klinikum München West, Munich, Germany
| | - Nekane Larburu
- Vicomtech, Health Research Institute, San Sebastian, Spain
| | - Iván Macía
- Vicomtech, Health Research Institute, San Sebastian, Spain
| | - Roma Maguire
- University of Strathclyde, Glasgow, United Kingdom
| | - Christos Maramis
- eHealth Lab, Institute of Applied Biosciences - Centre for Research & Technology Hellas, Thessaloniki, Greece
| | | | - Philip Morrow
- School of Computing, Ulster University, Newtownabbey, United Kingdom
| | - Naiara Muro
- Vicomtech, Health Research Institute, San Sebastian, Spain
| | | | - Andrik Rampun
- Academic Unit of Radiology, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | | | - Bryan Scotney
- School of Computing, Ulster University, Newtownabbey, United Kingdom
| | | | - Hui Wang
- School of Computing and Engineering, University of West London, London, United Kingdom
| | | | - Reyer Zwiggelaar
- Department of Computer Science, Aberystwyth University, Aberystwyth, United Kingdom
| |
Collapse
|
27
|
Caravagna G, Sanguinetti G, Graham TA, Sottoriva A. The MOBSTER R package for tumour subclonal deconvolution from bulk DNA whole-genome sequencing data. BMC Bioinformatics 2020; 21:531. [PMID: 33203356 PMCID: PMC7672894 DOI: 10.1186/s12859-020-03863-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/04/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The large-scale availability of whole-genome sequencing profiles from bulk DNA sequencing of cancer tissues is fueling the application of evolutionary theory to cancer. From a bulk biopsy, subclonal deconvolution methods are used to determine the composition of cancer subpopulations in the biopsy sample, a fundamental step to determine clonal expansions and their evolutionary trajectories. RESULTS In a recent work we have developed a new model-based approach to carry out subclonal deconvolution from the site frequency spectrum of somatic mutations. This new method integrates, for the first time, an explicit model for neutral evolutionary forces that participate in clonal expansions; in that work we have also shown that our method improves largely over competing data-driven methods. In this Software paper we present mobster, an open source R package built around our new deconvolution approach, which provides several functions to plot data and fit models, assess their confidence and compute further evolutionary analyses that relate to subclonal deconvolution. CONCLUSIONS We present the mobster package for tumour subclonal deconvolution from bulk sequencing, the first approach to integrate Machine Learning and Population Genetics which can explicitly model co-existing neutral and positive selection in cancer. We showcase the analysis of two datasets, one simulated and one from a breast cancer patient, and overview all package functionalities.
Collapse
Affiliation(s)
- Giulio Caravagna
- University of Trieste, Trieste, Italy.
- The Institute of Cancer Research, London, UK.
| | | | - Trevor A Graham
- Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | |
Collapse
|
28
|
Angaroni F, Graudenzi A, Rossignolo M, Maspero D, Calarco T, Piazza R, Montangero S, Antoniotti M. An Optimal Control Framework for the Automated Design of Personalized Cancer Treatments. Front Bioeng Biotechnol 2020; 8:523. [PMID: 32548108 PMCID: PMC7270334 DOI: 10.3389/fbioe.2020.00523] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
One of the key challenges in current cancer research is the development of computational strategies to support clinicians in the identification of successful personalized treatments. Control theory might be an effective approach to this end, as proven by the long-established application to therapy design and testing. In this respect, we here introduce the Control Theory for Therapy Design (CT4TD) framework, which employs optimal control theory on patient-specific pharmacokinetics (PK) and pharmacodynamics (PD) models, to deliver optimized therapeutic strategies. The definition of personalized PK/PD models allows to explicitly consider the physiological heterogeneity of individuals and to adapt the therapy accordingly, as opposed to standard clinical practices. CT4TD can be used in two distinct scenarios. At the time of the diagnosis, CT4TD allows to set optimized personalized administration strategies, aimed at reaching selected target drug concentrations, while minimizing the costs in terms of toxicity and adverse effects. Moreover, if longitudinal data on patients under treatment are available, our approach allows to adjust the ongoing therapy, by relying on simplified models of cancer population dynamics, with the goal of minimizing or controlling the tumor burden. CT4TD is highly scalable, as it employs the efficient dCRAB/RedCRAB optimization algorithm, and the results are robust, as proven by extensive tests on synthetic data. Furthermore, the theoretical framework is general, and it might be applied to any therapy for which a PK/PD model can be estimated, and for any kind of administration and cost. As a proof of principle, we present the application of CT4TD to Imatinib administration in Chronic Myeloid leukemia, in which we adopt a simplified model of cancer population dynamics. In particular, we show that the optimized therapeutic strategies are diversified among patients, and display improvements with respect to the current standard regime.
Collapse
Affiliation(s)
- Fabrizio Angaroni
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy
| | - Alex Graudenzi
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy
- Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
| | - Marco Rossignolo
- Center for Integrated Quantum Science and Technologies, Institute for Quantum Optics, Universitat Ulm, Ulm, Germany
- Istituto Nazionale di Fisica Nucleare (INFN), Padova, Italy
| | - Davide Maspero
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy
- Institute of Molecular Bioimaging and Physiology, Consiglio Nazionale delle Ricerche (IBFM-CNR), Segrate, Milan, Italy
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tommaso Calarco
- Forschungszentrum Jülich, Institute of Quantum Control (PGI-8), Jülich, Germany
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Hematology and Clinical Research Unit, San Gerardo Hospital, Monza, Italy
| | - Simone Montangero
- Istituto Nazionale di Fisica Nucleare (INFN), Padova, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padova, Padova, Italy
| | - Marco Antoniotti
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre - B4, Milan, Italy
| |
Collapse
|
29
|
Qian J, Zhao S, Zou Y, Rahman SMJ, Senosain MF, Stricker T, Chen H, Powell CA, Borczuk AC, Massion PP. Genomic Underpinnings of Tumor Behavior in In Situ and Early Lung Adenocarcinoma. Am J Respir Crit Care Med 2020; 201:697-706. [PMID: 31747302 PMCID: PMC7068818 DOI: 10.1164/rccm.201902-0294oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 11/19/2019] [Indexed: 01/15/2023] Open
Abstract
Rationale: We have a limited understanding of the molecular underpinnings of early adenocarcinoma (ADC) progression. We hypothesized that the behavior of early ADC can be predicted based on genomic determinants.Objectives: To identify genomic alterations associated with resected indolent and aggressive early lung ADCs.Methods: DNA was extracted from 21 ADCs in situ (AISs), 27 minimally invasive ADCs (MIAs), and 54 fully invasive ADCs. This DNA was subjected to deep next-generation sequencing and tested against a custom panel of 347 cancer genes.Measurements and Main Results: Sequencing data was analyzed for associations among tumor mutation burden, frequency of mutations or copy number alterations, mutation signatures, intratumor heterogeneity, pathway alterations, histology, and overall survival. We found that deleterious mutation burden was significantly greater in invasive ADC, whereas more copy number loss was observed in AIS and MIA. Intratumor heterogeneity establishes early, as in AIS. Twenty-one significantly mutated genes were shared among the groups. Mutation signature profiling did not vary significantly, although the APOBEC signature was associated with ADC and poor survival. Subclonal KRAS mutations and a gene signature consisting of PIK3CG, ATM, EPPK1, EP300, or KMT2C mutations were also associated with poor survival. Mutations of KRAS, TP53, and NF1 were found to increase in frequency from AIS and MIA to ADC. A cancer progression model revealed selective early and late drivers.Conclusions: Our results reveal several genetic driver events, clonality, and mutational signatures associated with poor outcome in early lung ADC, with potential future implications for the detection and management of ADC.
Collapse
Affiliation(s)
- Jun Qian
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Early Cancer Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center
- Center for Pecision Medicine, Department of Biomedical Informatics
| | | | - Yong Zou
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Early Cancer Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center
| | - S. M. Jamshedur Rahman
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Early Cancer Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center
| | - Maria-Fernanda Senosain
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Early Cancer Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center
| | - Thomas Stricker
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Heidi Chen
- Center for Pecision Medicine, Department of Biomedical Informatics
| | | | - Alain C. Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, New York
| | - Pierre P. Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Early Cancer Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center
| |
Collapse
|
30
|
Yu XJ, Chen G, Yang J, Yu GC, Zhu PF, Jiang ZK, Feng K, Lu Y, Bao B, Zhong FM. Smoking alters the evolutionary trajectory of non-small cell lung cancer. Exp Ther Med 2019; 18:3315-3324. [PMID: 31602204 PMCID: PMC6777332 DOI: 10.3892/etm.2019.7958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 05/16/2019] [Indexed: 12/14/2022] Open
Abstract
Smoking is the biggest risk factor for lung cancer. Smokers have a much higher chance of developing lung tumors with a worse survival rate; however, non-smokers also develop lung tumors. A number of questions remain including the underlying difference between smoker and non-smoker lung cancer patients and the involvement of genetic and epigenetic processes in tumor development. The present study analyzed the mutation data of 100 non-small cell lung cancer (NSCLC) patients, 12 non-smokers, 48 ex-smokers and 40 smokers, from Tracking Non-Small Cell Lung Cancer Evolution through Therapy Consortium. A total of 68 genes exhibited different mutation patterns across non-smokers, ex-smokers and smokers. A number of these 68 genes encode membrane proteins with biological regulation, metabolic process, and response to stimulus functions. For each group of patients, the top 10 most frequently mutated genes were selected and their oncogenetic tree inferred, which reflected how the genes evolve during tumor genesis. By comparing the oncogenetic trees of non-smokers and smokers, it was identified that in non-smokers, the mutation of epidermal growth factor receptor (EGFR) was an early genetic alteration event and EGFR was the key driver, but in smokers, the mutation of titin (TTN) was more important. Based on network analysis, TTN can interact with spectrin α erythrocytic 1 through calmodulin 2 and troponin C1. These genetic differences during tumorigenesis of non-smoker and smoker lung cancer patients provided novel insights into the effects of smoking on the evolutionary trajectory of non-small cell lung cancer and may prove helpful for targeted therapy of different lung cancer subtypes.
Collapse
Affiliation(s)
- Xiao-Jun Yu
- Department of Thoracic Surgery, The First People's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang 311400, P.R. China
| | - Gang Chen
- Department of Thoracic Surgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Jun Yang
- Department of Thoracic Surgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Guo-Can Yu
- Department of Thoracic Surgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Peng-Fei Zhu
- Department of Thoracic Surgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Zheng-Ke Jiang
- Department of Surgery, Hangzhou Fuyang Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang 311400, P.R. China
| | - Kan Feng
- Department of Thoracic Surgery, The First People's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang 311400, P.R. China
| | - Yong Lu
- Department of Thoracic Surgery, The First People's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang 311400, P.R. China
| | - Bin Bao
- Department of Thoracic Surgery, The First People's Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang 311400, P.R. China
| | - Fang-Ming Zhong
- Department of Thoracic Surgery, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
31
|
Diaz-Uriarte R, Vasallo C. Every which way? On predicting tumor evolution using cancer progression models. PLoS Comput Biol 2019; 15:e1007246. [PMID: 31374072 PMCID: PMC6693785 DOI: 10.1371/journal.pcbi.1007246] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 08/14/2019] [Accepted: 07/05/2019] [Indexed: 11/18/2022] Open
Abstract
Successful prediction of the likely paths of tumor progression is valuable for diagnostic, prognostic, and treatment purposes. Cancer progression models (CPMs) use cross-sectional samples to identify restrictions in the order of accumulation of driver mutations and thus CPMs encode the paths of tumor progression. Here we analyze the performance of four CPMs to examine whether they can be used to predict the true distribution of paths of tumor progression and to estimate evolutionary unpredictability. Employing simulations we show that if fitness landscapes are single peaked (have a single fitness maximum) there is good agreement between true and predicted distributions of paths of tumor progression when sample sizes are large, but performance is poor with the currently common much smaller sample sizes. Under multi-peaked fitness landscapes (i.e., those with multiple fitness maxima), performance is poor and improves only slightly with sample size. In all cases, detection regime (when tumors are sampled) is a key determinant of performance. Estimates of evolutionary unpredictability from the best performing CPM, among the four examined, tend to overestimate the true unpredictability and the bias is affected by detection regime; CPMs could be useful for estimating upper bounds to the true evolutionary unpredictability. Analysis of twenty-two cancer data sets shows low evolutionary unpredictability for several of the data sets. But most of the predictions of paths of tumor progression are very unreliable, and unreliability increases with the number of features analyzed. Our results indicate that CPMs could be valuable tools for predicting cancer progression but that, currently, obtaining useful predictions of paths of tumor progression from CPMs is dubious, and emphasize the need for methodological work that can account for the probably multi-peaked fitness landscapes in cancer.
Collapse
Affiliation(s)
- Ramon Diaz-Uriarte
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (UAM-CSIC), Madrid, Spain
| | - Claudia Vasallo
- Department of Biochemistry, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (UAM-CSIC), Madrid, Spain
| |
Collapse
|
32
|
Ramazzotti D, Graudenzi A, De Sano L, Antoniotti M, Caravagna G. Learning mutational graphs of individual tumour evolution from single-cell and multi-region sequencing data. BMC Bioinformatics 2019; 20:210. [PMID: 31023236 PMCID: PMC6485126 DOI: 10.1186/s12859-019-2795-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 04/08/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND A large number of algorithms is being developed to reconstruct evolutionary models of individual tumours from genome sequencing data. Most methods can analyze multiple samples collected either through bulk multi-region sequencing experiments or the sequencing of individual cancer cells. However, rarely the same method can support both data types. RESULTS We introduce TRaIT, a computational framework to infer mutational graphs that model the accumulation of multiple types of somatic alterations driving tumour evolution. Compared to other tools, TRaIT supports multi-region and single-cell sequencing data within the same statistical framework, and delivers expressive models that capture many complex evolutionary phenomena. TRaIT improves accuracy, robustness to data-specific errors and computational complexity compared to competing methods. CONCLUSIONS We show that the application of TRaIT to single-cell and multi-region cancer datasets can produce accurate and reliable models of single-tumour evolution, quantify the extent of intra-tumour heterogeneity and generate new testable experimental hypotheses.
Collapse
Affiliation(s)
| | - Alex Graudenzi
- Dipartimento di Informatica, Sistemistica e Comunicazione, Università degli Studi di Milano-Bicocca, Viale Sarca 336, Milan, 20126 Italy
- Institute of Molecular Bioimaging and Physiology of the Italian National Research Council (IBFM-CNR), Viale F.lli Cervi 93, Segrate, Milan, 20090 Italy
| | - Luca De Sano
- Dipartimento di Informatica, Sistemistica e Comunicazione, Università degli Studi di Milano-Bicocca, Viale Sarca 336, Milan, 20126 Italy
| | - Marco Antoniotti
- Dipartimento di Informatica, Sistemistica e Comunicazione, Università degli Studi di Milano-Bicocca, Viale Sarca 336, Milan, 20126 Italy
- Milan Center for Neuroscience, Università degli Studi di Milano-Bicocca, San Gerardo Hospital, Via Pergolesi 33, Monza, 20052 Italy
| | - Giulio Caravagna
- Centre for Evolution and Cancer, The Institute of Cancer Research, 15 Cotswold Road, London, SM2 5NG UK
| |
Collapse
|
33
|
Diaz-Uriarte R. Cancer progression models and fitness landscapes: a many-to-many relationship. Bioinformatics 2018; 34:836-844. [PMID: 29048486 PMCID: PMC6031050 DOI: 10.1093/bioinformatics/btx663] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/17/2017] [Indexed: 11/13/2022] Open
Abstract
Motivation The identification of constraints, due to gene interactions, in the order of accumulation of mutations during cancer progression can allow us to single out therapeutic targets. Cancer progression models (CPMs) use genotype frequency data from cross-sectional samples to identify these constraints, and return Directed Acyclic Graphs (DAGs) of restrictions where arrows indicate dependencies or constraints. On the other hand, fitness landscapes, which map genotypes to fitness, contain all possible paths of tumor progression. Thus, we expect a correspondence between DAGs from CPMs and the fitness landscapes where evolution happened. But many fitness landscapes-e.g. those with reciprocal sign epistasis-cannot be represented by CPMs. Results Using simulated data under 500 fitness landscapes, I show that CPMs' performance (prediction of genotypes that can exist) degrades with reciprocal sign epistasis. There is large variability in the DAGs inferred from each landscape, which is also affected by mutation rate, detection regime and fitness landscape features, in ways that depend on CPM method. Using three cancer datasets, I show that these problems strongly affect the analysis of empirical data: fitness landscapes that are widely different from each other produce data similar to the empirically observed ones and lead to DAGs that infer very different restrictions. Because reciprocal sign epistasis can be common in cancer, these results question the use and interpretation of CPMs. Availability and implementation Code available from Supplementary Material. Contact ramon.diaz@iib.uam.es. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ramon Diaz-Uriarte
- Department of Biochemistry, Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas "Alberto Sols" (UAM-CSIC), Madrid 28029, Spain
| |
Collapse
|
34
|
Graudenzi A, Maspero D, Di Filippo M, Gnugnoli M, Isella C, Mauri G, Medico E, Antoniotti M, Damiani C. Integration of transcriptomic data and metabolic networks in cancer samples reveals highly significant prognostic power. J Biomed Inform 2018; 87:37-49. [PMID: 30244122 DOI: 10.1016/j.jbi.2018.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/07/2018] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
Abstract
Effective stratification of cancer patients on the basis of their molecular make-up is a key open challenge. Given the altered and heterogenous nature of cancer metabolism, we here propose to use the overall expression of central carbon metabolism as biomarker to characterize groups of patients with important characteristics, such as response to ad-hoc therapeutic strategies and survival expectancy. To this end, we here introduce the data integration framework named Metabolic Reaction Enrichment Analysis (MaREA), which strives to characterize the metabolic deregulations that distinguish cancer phenotypes, by projecting RNA-seq data onto metabolic networks, without requiring metabolic measurements. MaREA computes a score for each network reaction, based on the expression of the set of genes encoding for the associated enzyme(s). The scores are first used as features for cluster analysis and then to rank and visualize in an organized fashion the metabolic deregulations that distinguish cancer sub-types. We applied our method to recent lung and breast cancer RNA-seq datasets from The Cancer Genome Atlas and we were able to identify subgroups of patients with significant differences in survival expectancy. We show how the prognostic power of MaREA improves when an extracted and further curated core model focusing on central carbon metabolism is used rather than the genome-wide reference network. The visualization of the metabolic differences between the groups with best and worst prognosis allowed to identify and analyze key metabolic properties related to cancer aggressiveness. Some of these properties are shared across different cancer (sub) types, e.g., the up-regulation of nucleic acid and amino acid synthesis, whereas some other appear to be tumor-specific, such as the up- or down-regulation of the phosphoenolpyruvate carboxykinase reaction, which display different patterns in distinct tumor (sub)types. These results might be soon employed to deliver highly automated diagnostic and prognostic strategies for cancer patients.
Collapse
Affiliation(s)
- Alex Graudenzi
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy
| | - Davide Maspero
- Department of Biotechnology and Biosciences, University Milano-Bicocca, Milan, Italy
| | - Marzia Di Filippo
- Department of Biotechnology and Biosciences, University Milano-Bicocca, Milan, Italy; SYSBIO Centre of Systems Biology, University Milano-Bicocca, Milan, Italy
| | - Marco Gnugnoli
- Department of Biotechnology and Biosciences, University Milano-Bicocca, Milan, Italy; SYSBIO Centre of Systems Biology, University Milano-Bicocca, Milan, Italy
| | - Claudio Isella
- University of Torino, Department of Oncology, Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Giancarlo Mauri
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy; SYSBIO Centre of Systems Biology, University Milano-Bicocca, Milan, Italy
| | - Enzo Medico
- University of Torino, Department of Oncology, Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Marco Antoniotti
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy; Milan Center for Neuroscience, University of Milan-Bicocca, Monza, Italy
| | - Chiara Damiani
- Department of Informatics, Systems and Communication, University of Milan-Bicocca, Milan, Italy; SYSBIO Centre of Systems Biology, University Milano-Bicocca, Milan, Italy.
| |
Collapse
|
35
|
Caravagna G, Giarratano Y, Ramazzotti D, Tomlinson I, Graham TA, Sanguinetti G, Sottoriva A. Detecting repeated cancer evolution from multi-region tumor sequencing data. Nat Methods 2018; 15:707-714. [PMID: 30171232 PMCID: PMC6380470 DOI: 10.1038/s41592-018-0108-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 07/23/2018] [Indexed: 12/13/2022]
Abstract
Recurrent successions of genomic changes, both within and between patients, reflect repeated evolutionary processes that are valuable for the anticipation of cancer progression. Multi-region sequencing allows the temporal order of some genomic changes in a tumor to be inferred, but the robust identification of repeated evolution across patients remains a challenge. We developed a machine-learning method based on transfer learning that allowed us to overcome the stochastic effects of cancer evolution and noise in data and identified hidden evolutionary patterns in cancer cohorts. When applied to multi-region sequencing datasets from lung, breast, renal, and colorectal cancer (768 samples from 178 patients), our method detected repeated evolutionary trajectories in subgroups of patients, which were reproduced in single-sample cohorts (n = 2,935). Our method provides a means of classifying patients on the basis of how their tumor evolved, with implications for the anticipation of disease progression.
Collapse
Affiliation(s)
- Giulio Caravagna
- Evolutionary Genomics and Modelling Lab, Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
- School of Informatics, University of Edinburgh, Edinburgh, UK.
| | - Ylenia Giarratano
- School of Informatics, University of Edinburgh, Edinburgh, UK
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, UK
| | | | - Ian Tomlinson
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Trevor A Graham
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Andrea Sottoriva
- Evolutionary Genomics and Modelling Lab, Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
36
|
Gambacorti-Passerini C, Piazza R. How "precise" is precision medicine in hematology? Haematologica 2018; 102:4-6. [PMID: 28040787 DOI: 10.3324/haematol.2016.155267] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
| | - Rocco Piazza
- Hematology, School of Medicine and Surgery, University of Milano Bicocca, via Cadore 48, Monza, Italy
| |
Collapse
|
37
|
Ramazzotti D, Graudenzi A, Caravagna G, Antoniotti M. Modeling Cumulative Biological Phenomena with Suppes-Bayes Causal Networks. Evol Bioinform Online 2018; 14:1176934318785167. [PMID: 30013303 PMCID: PMC6043942 DOI: 10.1177/1176934318785167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/27/2018] [Indexed: 12/18/2022] Open
Abstract
Several diseases related to cell proliferation are characterized by the accumulation of somatic DNA changes, with respect to wild-type conditions. Cancer and HIV are 2 common examples of such diseases, where the mutational load in the cancerous/viral population increases over time. In these cases, selective pressures are often observed along with competition, co-operation, and parasitism among distinct cellular clones. Recently, we presented a mathematical framework to model these phenomena, based on a combination of Bayesian inference and Suppes’ theory of probabilistic causation, depicted in graphical structures dubbed Suppes-Bayes Causal Networks (SBCNs). The SBCNs are generative probabilistic graphical models that recapitulate the potential ordering of accumulation of such DNA changes during the progression of the disease. Such models can be inferred from data by exploiting likelihood-based model selection strategies with regularization. In this article, we discuss the theoretical foundations of our approach and we investigate in depth the influence on the model selection task of (1) the poset based on Suppes’ theory and (2) different regularization strategies. Furthermore, we provide an example of application of our framework to HIV genetic data highlighting the valuable insights provided by the inferred SBCN
Collapse
Affiliation(s)
| | - Alex Graudenzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | | | - Marco Antoniotti
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
38
|
Sun Y, Yao J, Yang L, Chen R, Nowak NJ, Goodison S. Computational approach for deriving cancer progression roadmaps from static sample data. Nucleic Acids Res 2017; 45:e69. [PMID: 28108658 PMCID: PMC5436003 DOI: 10.1093/nar/gkx003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/07/2017] [Indexed: 12/26/2022] Open
Abstract
As with any biological process, cancer development is inherently dynamic. While major efforts continue to catalog the genomic events associated with human cancer, it remains difficult to interpret and extrapolate the accumulating data to provide insights into the dynamic aspects of the disease. Here, we present a computational strategy that enables the construction of a cancer progression model using static tumor sample data. The developed approach overcame many technical limitations of existing methods. Application of the approach to breast cancer data revealed a linear, branching model with two distinct trajectories for malignant progression. The validity of the constructed model was demonstrated in 27 independent breast cancer data sets, and through visualization of the data in the context of disease progression we were able to identify a number of potentially key molecular events in the advance of breast cancer to malignancy.
Collapse
Affiliation(s)
- Yijun Sun
- Department of Microbiology and Immunology.,Department of Computer Science and Engineering.,Department of Biostatistics, The State University of New York, Buffalo, NY14203, USA.,Department of Biochemistry The State University of New York, Buffalo, NY14203, USA
| | - Jin Yao
- Department of Microbiology and Immunology
| | - Le Yang
- Department of Computer Science and Engineering
| | - Runpu Chen
- Department of Computer Science and Engineering
| | - Norma J Nowak
- Department of Bioinformatics and Biostatistics Roswell Park Cancer Institute, Buffalo, NY 14201, USA
| | - Steve Goodison
- Department of Health Sciences Research Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
39
|
Abstract
Rapid advances in high-throughput sequencing and a growing realization of the importance of evolutionary theory to cancer genomics have led to a proliferation of phylogenetic studies of tumour progression. These studies have yielded not only new insights but also a plethora of experimental approaches, sometimes reaching conflicting or poorly supported conclusions. Here, we consider this body of work in light of the key computational principles underpinning phylogenetic inference, with the goal of providing practical guidance on the design and analysis of scientifically rigorous tumour phylogeny studies. We survey the range of methods and tools available to the researcher, their key applications, and the various unsolved problems, closing with a perspective on the prospects and broader implications of this field.
Collapse
Affiliation(s)
- Russell Schwartz
- Department of Biological Sciences and Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania 15217, USA
| | - Alejandro A Schäffer
- Computational Biology Branch, National Center for Biotechnology Information, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
40
|
Pirkl M, Diekmann M, van der Wees M, Beerenwinkel N, Fröhlich H, Markowetz F. Inferring modulators of genetic interactions with epistatic nested effects models. PLoS Comput Biol 2017; 13:e1005496. [PMID: 28406896 PMCID: PMC5407847 DOI: 10.1371/journal.pcbi.1005496] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 04/27/2017] [Accepted: 04/03/2017] [Indexed: 12/27/2022] Open
Abstract
Maps of genetic interactions can dissect functional redundancies in cellular networks. Gene expression profiles as high-dimensional molecular readouts of combinatorial perturbations provide a detailed view of genetic interactions, but can be hard to interpret if different gene sets respond in different ways (called mixed epistasis). Here we test the hypothesis that mixed epistasis between a gene pair can be explained by the action of a third gene that modulates the interaction. We have extended the framework of Nested Effects Models (NEMs), a type of graphical model specifically tailored to analyze high-dimensional gene perturbation data, to incorporate logical functions that describe interactions between regulators on downstream genes and proteins. We benchmark our approach in the controlled setting of a simulation study and show high accuracy in inferring the correct model. In an application to data from deletion mutants of kinases and phosphatases in S. cerevisiae we show that epistatic NEMs can point to modulators of genetic interactions. Our approach is implemented in the R-package 'epiNEM' available from https://github.com/cbg-ethz/epiNEM and https://bioconductor.org/packages/epiNEM/.
Collapse
Affiliation(s)
- Martin Pirkl
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Madeline Diekmann
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Niko Beerenwinkel
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Holger Fröhlich
- Bonn-Aachen International Center for IT (B-IT), University of Bonn, Bonn, Germany
- UCB Biosciences GmbH, Monheim, Germany
| | - Florian Markowetz
- University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| |
Collapse
|
41
|
Peterson LE, Kovyrshina T. Progression inference for somatic mutations in cancer. Heliyon 2017; 3:e00277. [PMID: 28492066 PMCID: PMC5415494 DOI: 10.1016/j.heliyon.2017.e00277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/08/2017] [Accepted: 03/23/2017] [Indexed: 01/05/2023] Open
Abstract
Computational methods were employed to determine progression inference of genomic alterations in commonly occurring cancers. Using cross-sectional TCGA data, we computed evolutionary trajectories involving selectivity relationships among pairs of gene-specific genomic alterations such as somatic mutations, deletions, amplifications, downregulation, and upregulation among the top 20 driver genes associated with each cancer. Results indicate that the majority of hierarchies involved TP53, PIK3CA, ERBB2, APC, KRAS, EGFR, IDH1, VHL, etc. Research into the order and accumulation of genomic alterations among cancer driver genes will ever-increase as the costs of nextgen sequencing subside, and personalized/precision medicine incorporates whole-genome scans into the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Leif E. Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Dept. of Healthcare Policy and Research, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
- Dept. of Biostatistics, School of Public Health, University of Texas – Health Science Center, Houston, TX 77030, USA
- Dept. of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dept. of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Tatiana Kovyrshina
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Dept. of Mathematics and Statistics, University of Houston – Downtown, Houston, TX 77002, USA
| |
Collapse
|