1
|
Kemkar S, Tao M, Ghosh A, Stamatakos G, Graf N, Poorey K, Balakrishnan U, Trask N, Radhakrishnan R. Towards verifiable cancer digital twins: tissue level modeling protocol for precision medicine. Front Physiol 2024; 15:1473125. [PMID: 39507514 PMCID: PMC11537925 DOI: 10.3389/fphys.2024.1473125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Cancer exhibits substantial heterogeneity, manifesting as distinct morphological and molecular variations across tumors, which frequently undermines the efficacy of conventional oncological treatments. Developments in multiomics and sequencing technologies have paved the way for unraveling this heterogeneity. Nevertheless, the complexity of the data gathered from these methods cannot be fully interpreted through multimodal data analysis alone. Mathematical modeling plays a crucial role in delineating the underlying mechanisms to explain sources of heterogeneity using patient-specific data. Intra-tumoral diversity necessitates the development of precision oncology therapies utilizing multiphysics, multiscale mathematical models for cancer. This review discusses recent advancements in computational methodologies for precision oncology, highlighting the potential of cancer digital twins to enhance patient-specific decision-making in clinical settings. We review computational efforts in building patient-informed cellular and tissue-level models for cancer and propose a computational framework that utilizes agent-based modeling as an effective conduit to integrate cancer systems models that encode signaling at the cellular scale with digital twin models that predict tissue-level response in a tumor microenvironment customized to patient information. Furthermore, we discuss machine learning approaches to building surrogates for these complex mathematical models. These surrogates can potentially be used to conduct sensitivity analysis, verification, validation, and uncertainty quantification, which is especially important for tumor studies due to their dynamic nature.
Collapse
Affiliation(s)
- Sharvari Kemkar
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Mengdi Tao
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alokendra Ghosh
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Georgios Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Zografos, Greece
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Kunal Poorey
- Department of Systems Biology, Sandia National Laboratories, Livermore, CA, United States
| | - Uma Balakrishnan
- Department of Quant Modeling and SW Eng, Sandia National Laboratories, Livermore, CA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Nathaniel Trask
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, United States
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
2
|
Ma C, Gurkan-Cavusoglu E. A comprehensive review of computational cell cycle models in guiding cancer treatment strategies. NPJ Syst Biol Appl 2024; 10:71. [PMID: 38969664 PMCID: PMC11226463 DOI: 10.1038/s41540-024-00397-7] [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: 01/26/2024] [Accepted: 06/24/2024] [Indexed: 07/07/2024] Open
Abstract
This article reviews the current knowledge and recent advancements in computational modeling of the cell cycle. It offers a comparative analysis of various modeling paradigms, highlighting their unique strengths, limitations, and applications. Specifically, the article compares deterministic and stochastic models, single-cell versus population models, and mechanistic versus abstract models. This detailed analysis helps determine the most suitable modeling framework for various research needs. Additionally, the discussion extends to the utilization of these computational models to illuminate cell cycle dynamics, with a particular focus on cell cycle viability, crosstalk with signaling pathways, tumor microenvironment, DNA replication, and repair mechanisms, underscoring their critical roles in tumor progression and the optimization of cancer therapies. By applying these models to crucial aspects of cancer therapy planning for better outcomes, including drug efficacy quantification, drug discovery, drug resistance analysis, and dose optimization, the review highlights the significant potential of computational insights in enhancing the precision and effectiveness of cancer treatments. This emphasis on the intricate relationship between computational modeling and therapeutic strategy development underscores the pivotal role of advanced modeling techniques in navigating the complexities of cell cycle dynamics and their implications for cancer therapy.
Collapse
Affiliation(s)
- Chenhui Ma
- Department of Electrical, Computer and Systems Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Evren Gurkan-Cavusoglu
- Department of Electrical, Computer and Systems Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
3
|
Kolokotroni E, Abler D, Ghosh A, Tzamali E, Grogan J, Georgiadi E, Büchler P, Radhakrishnan R, Byrne H, Sakkalis V, Nikiforaki K, Karatzanis I, McFarlane NJB, Kaba D, Dong F, Bohle RM, Meese E, Graf N, Stamatakos G. A Multidisciplinary Hyper-Modeling Scheme in Personalized In Silico Oncology: Coupling Cell Kinetics with Metabolism, Signaling Networks, and Biomechanics as Plug-In Component Models of a Cancer Digital Twin. J Pers Med 2024; 14:475. [PMID: 38793058 PMCID: PMC11122096 DOI: 10.3390/jpm14050475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/11/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024] Open
Abstract
The massive amount of human biological, imaging, and clinical data produced by multiple and diverse sources necessitates integrative modeling approaches able to summarize all this information into answers to specific clinical questions. In this paper, we present a hypermodeling scheme able to combine models of diverse cancer aspects regardless of their underlying method or scale. Describing tissue-scale cancer cell proliferation, biomechanical tumor growth, nutrient transport, genomic-scale aberrant cancer cell metabolism, and cell-signaling pathways that regulate the cellular response to therapy, the hypermodel integrates mutation, miRNA expression, imaging, and clinical data. The constituting hypomodels, as well as their orchestration and links, are described. Two specific cancer types, Wilms tumor (nephroblastoma) and non-small cell lung cancer, are addressed as proof-of-concept study cases. Personalized simulations of the actual anatomy of a patient have been conducted. The hypermodel has also been applied to predict tumor control after radiotherapy and the relationship between tumor proliferative activity and response to neoadjuvant chemotherapy. Our innovative hypermodel holds promise as a digital twin-based clinical decision support system and as the core of future in silico trial platforms, although additional retrospective adaptation and validation are necessary.
Collapse
Affiliation(s)
- Eleni Kolokotroni
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, 157 80 Zografos, Greece;
| | - Daniel Abler
- Department of Oncology, Geneva University Hospitals and University of Geneva, 1205 Geneva, Switzerland;
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Alokendra Ghosh
- Department of Chemical and Biomolecular Engineering, Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.); (R.R.)
| | - Eleftheria Tzamali
- Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (V.S.); (K.N.); (I.K.)
| | - James Grogan
- Irish Centre for High End Computing, University of Galway, H91 TK33 Galway, Ireland;
| | - Eleni Georgiadi
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, 157 80 Zografos, Greece;
- Biomedical Engineering Department, University of West Attica, 12243 Egaleo, Greece
| | | | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.); (R.R.)
| | - Helen Byrne
- Mathematical Institute, University of Oxford, Oxford OX1 2JD, UK;
| | - Vangelis Sakkalis
- Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (V.S.); (K.N.); (I.K.)
| | - Katerina Nikiforaki
- Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (V.S.); (K.N.); (I.K.)
| | - Ioannis Karatzanis
- Institute of Computer Science, Foundation for Research and Technology—Hellas, 70013 Heraklion, Greece; (E.T.); (V.S.); (K.N.); (I.K.)
| | | | - Djibril Kaba
- Department of Computer Science and Technology, University of Bedfordshire, Luton LU1 3JU, UK;
| | - Feng Dong
- Department of Computer & Information Sciences, University of Strathclyde, Glasgow G1 1XH, UK;
| | - Rainer M. Bohle
- Department of Pathology, Saarland University, 66421 Homburg, Germany;
| | - Eckart Meese
- Department of Human Genetics, Saarland University, 66421 Homburg, Germany;
| | - Norbert Graf
- Department of Paediatric Oncology and Haematology, Saarland University, 66421 Homburg, Germany;
| | - Georgios Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, 157 80 Zografos, Greece;
| |
Collapse
|
4
|
Gondal MN, Chaudhary SU. Navigating Multi-Scale Cancer Systems Biology Towards Model-Driven Clinical Oncology and Its Applications in Personalized Therapeutics. Front Oncol 2021; 11:712505. [PMID: 34900668 PMCID: PMC8652070 DOI: 10.3389/fonc.2021.712505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022] Open
Abstract
Rapid advancements in high-throughput omics technologies and experimental protocols have led to the generation of vast amounts of scale-specific biomolecular data on cancer that now populates several online databases and resources. Cancer systems biology models built using this data have the potential to provide specific insights into complex multifactorial aberrations underpinning tumor initiation, development, and metastasis. Furthermore, the annotation of these single- and multi-scale models with patient data can additionally assist in designing personalized therapeutic interventions as well as aid in clinical decision-making. Here, we have systematically reviewed the emergence and evolution of (i) repositories with scale-specific and multi-scale biomolecular cancer data, (ii) systems biology models developed using this data, (iii) associated simulation software for the development of personalized cancer therapeutics, and (iv) translational attempts to pipeline multi-scale panomics data for data-driven in silico clinical oncology. The review concludes that the absence of a generic, zero-code, panomics-based multi-scale modeling pipeline and associated software framework, impedes the development and seamless deployment of personalized in silico multi-scale models in clinical settings.
Collapse
Affiliation(s)
- Mahnoor Naseer Gondal
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Safee Ullah Chaudhary
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
5
|
Lin C, Zhu X, Jin Q, Sui A, Li J, Shen L. Effects of Holothurian Glycosaminoglycan on the Sensitivity of Lung Cancer to Chemotherapy. Integr Cancer Ther 2021; 19:1534735420911430. [PMID: 32202167 PMCID: PMC7092648 DOI: 10.1177/1534735420911430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sea cucumber is a kind of food. Holothurian glycosaminoglycan (hGAG) is extracted from the body wall of the sea cucumber. Administration of hGAG and cisplatin (DDP) together to treat lung cancer was investigated. Lung adenocarcinoma A549 cells were cultured and divided into 4 groups: control group, hGAG 100 µg/mL group, DDP 3 µg/mL group, and hGAG 100 µg/mL + DDP 3 µg/mL group. Cell inhibition and apoptosis was evaluated by CCK8 and Hoechst33258 staining. Cell cycle was tested by Annexin V-FITC/PI (propidium iodide) double-staining and flow cytometry. The expression of mRNA and protein of Bcl-2, Bax, caspase-3, and survivin were detected by reverse transcriptase-polymerase chain reaction and Western blot, respectively. The results showed that hGAG combined with DDP enhanced the inhibitory effect of DDP on A549 lung cells through apoptosis pathway. The mechanism of apoptosis may be related to the reduction of Bcl-2 and survivin, as well as the ascension of Bax and caspase-3. hGAG could promote A549 cell cycle arrest in G1 and G2 phase and improve the DDP chemotherapy effects on A549 cells.
Collapse
Affiliation(s)
- Cunzhi Lin
- Department of Pulmonary Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xinhong Zhu
- Internal Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Qing Jin
- Department of Intensive Care Unit, The 903rd Hospital of People's Liberation Army, Hangzhou, Zhejiang, China
| | - Aihua Sui
- Department of Respiratory & Critical Care Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jinfeng Li
- Department of Respiratory & Critical Care Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Liyan Shen
- Department of Respiratory & Critical Care Medicine, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
6
|
Jin Q, Lin C, Zhu X, Cao Y, Guo C, Wang L. 125I seeds irradiation inhibits tumor growth and induces apoptosis by Ki-67, P21, survivin, livin and caspase-9 expression in lung carcinoma xenografts. Radiat Oncol 2020; 15:238. [PMID: 33059701 PMCID: PMC7559445 DOI: 10.1186/s13014-020-01682-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/06/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Lung cancer is a fatal disease and a serious health problem worldwide. Patients are usually diagnosed at an advanced stage, and the effectiveness of chemotherapy for such patients is very limited. Iodine 125 seed (125I) irradiation can be used as an important adjuvant treatment for lung carcinoma. The purpose of this study was to examine the role of irradiation by 125I seeds in human lung cancer xenograft model and to determine the underlying mechanisms involved, with a focus on apoptosis. METHODS 40 mice with A549 lung adenocarcinoma xenografts were randomly divided into 4 groups: control group (n = 10), sham seed (0 mCi) implant group (n = 10), 125I seed (0.6 mCi) implant group (n = 10) and 125I seed (0.8 mCi) implant group (n = 10), respectively. The body weight and tumor volume, were recorded every 4 days until the end of the study. Apoptotic cells were checked by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and activities of caspase-3 and caspase-8 enzyme were tested. Expression of P21, survivin, livin, caspase-9 and proliferating cell nuclear antigen (Ki-67) was detected with immunohistochemical staining. RESULTS The results of TUNEL staining assays showed that 125I seed irradiation suppresses the growth of lung cancer xenografts in nude mice and induced apoptosis. The activity of caspase-3 and caspase-8 was significantly higher. The expression levels Ki67, survivin and livin were substantially downregulated, while P21 and caspase-9 protein expression were significantly increased following 125I seed irradiation. This study revealed that 125I seed irradiation could significantly change apoptosis-related protein in human lung cancer xenografts. CONCLUSIONS Overall, our study demonstrates that radiation exposure by 125I seeds could be a new treatment option for lung cancer.
Collapse
Affiliation(s)
- Qing Jin
- Department of Critical Care Medicine, The 903th Hospital of PLA Joint Logistics Support Force, Zhejiang Province, Hangzhou, 310013, China
| | - Cunzhi Lin
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Xinhong Zhu
- Department of Internal Medicine, Qingdao Municipal Hospital, Qingdao, 266071, Shandong Province, China
| | - Yiwei Cao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Caihong Guo
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Lijun Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| |
Collapse
|
7
|
Kozłowska E, Suwiński R, Giglok M, Świerniak A, Kimmel M. Mathematical model predicts response to chemotherapy in advanced non-resectable non-small cell lung cancer patients treated with platinum-based doublet. PLoS Comput Biol 2020; 16:e1008234. [PMID: 33017420 PMCID: PMC7561182 DOI: 10.1371/journal.pcbi.1008234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 10/15/2020] [Accepted: 08/10/2020] [Indexed: 11/23/2022] Open
Abstract
We developed a computational platform including machine learning and a mechanistic mathematical model to find the optimal protocol for administration of platinum-doublet chemotherapy in a palliative setting. The platform has been applied to advanced metastatic non-small cell lung cancer (NSCLC). The 42 NSCLC patients treated with palliative intent at Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, were collected from a retrospective cohort of patients diagnosed in 2004–2014. Patients were followed-up, for three years. Clinical data collected include complete information about the clinical course of the patients including treatment schedule, response according to RECIST classification, and survival. The core of the platform is the mathematical model, in the form of a system of ordinary differential equations, describing dynamics of platinum-sensitive and platinum-resistant cancer cells and interactions reflecting competition for space and resources. The model is simulated stochastically by sampling the parameter values from a joint probability distribution function. The machine learning model is applied to calibrate the mathematical model and to fit it to the overall survival curve. The model simulations faithfully reproduce the clinical cohort at three levels long-term response (OS), the initial response (according to RECIST criteria), and the relationship between the number of chemotherapy cycles and time between two consecutive chemotherapy cycles. In addition, we investigated the relationship between initial and long-term response. We showed that those two variables do not correlate which means that we cannot predict patient survival solely based on the initial response. We also tested several chemotherapy schedules to find the best one for patients treated with palliative intent. We found that the optimal treatment schedule depends, among others, on the strength of competition among various subclones in a tumor. The computational platform developed allows optimizing chemotherapy protocols, within admissible limits of toxicity, for palliative treatment of metastatic NSCLC. The simplicity of the method allows its application to chemotherapy optimization in different cancers. Lung cancer is usually diagnosed at an advanced stage because of non-specific symptoms. The most common subtype of lung cancer is non-small cell lung cancer, which constitutes 80% of lung cancer cases. Here, we developed the methodology for finding the optimal treatment schedule for patients treated with palliative intent. The goal is not to cure the patients who are at an advanced stage but to prolong their survival by the administration of platinum-based chemotherapy. The method is based on the mathematical model describing the growth of tumors and its response to chemotherapy which is calibrated using real clinical data.
Collapse
Affiliation(s)
- Emilia Kozłowska
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka Gliwice, Poland
- * E-mail:
| | - Rafał Suwiński
- The 2 Radiotherapy and Chemotherapy Clinic, M. Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Monika Giglok
- The 2 Radiotherapy and Chemotherapy Clinic, M. Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Andrzej Świerniak
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka Gliwice, Poland
| | - Marek Kimmel
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka Gliwice, Poland
- Departments of Statistics and Bioengineering, Rice University, Houston Texas, United States of America
| |
Collapse
|
8
|
From tumour perfusion to drug delivery and clinical translation of in silico cancer models. Methods 2020; 185:82-93. [PMID: 32147442 DOI: 10.1016/j.ymeth.2020.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 02/13/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
In silico cancer models have demonstrated great potential as a tool to improve drug design, optimise the delivery of drugs to target sites in the host tissue and, hence, improve therapeutic efficacy and patient outcome. However, there are significant barriers to the successful translation of in silico technology from bench to bedside. More precisely, the specification of unknown model parameters, the necessity for models to adequately reflect in vivo conditions, and the limited amount of pertinent validation data to evaluate models' accuracy and assess their reliability, pose major obstacles in the path towards their clinical translation. This review aims to capture the state-of-the-art in in silico cancer modelling of vascularised solid tumour growth, and identify the important advances and barriers to success of these models in clinical oncology. Particular emphasis has been put on continuum-based models of cancer since they - amongst the class of mechanistic spatio-temporal modelling approaches - are well-established in simulating transport phenomena and the biomechanics of tissues, and have demonstrated potential for clinical translation. Three important avenues in in silico modelling are considered in this contribution: first, since systemic therapy is a major cancer treatment approach, we start with an overview of the tumour perfusion and angiogenesis in silico models. Next, we present the state-of-the-art in silico work encompassing the delivery of chemotherapeutic agents to cancer nanomedicines through the bloodstream, and then review continuum-based modelling approaches that demonstrate great promise for successful clinical translation. We conclude with a discussion of what we view to be the key challenges and opportunities for in silico modelling in personalised and precision medicine.
Collapse
|
9
|
Antonopoulos M, Dionysiou D, Stamatakos G, Uzunoglu N. Three-dimensional tumor growth in time-varying chemical fields: a modeling framework and theoretical study. BMC Bioinformatics 2019; 20:442. [PMID: 31455206 PMCID: PMC6712764 DOI: 10.1186/s12859-019-2997-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/16/2019] [Indexed: 01/10/2023] Open
Abstract
Background Contemporary biological observations have revealed a large variety of mechanisms acting during the expansion of a tumor. However, there are still many qualitative and quantitative aspects of the phenomenon that remain largely unknown. In this context, mathematical and computational modeling appears as an invaluable tool providing the means for conducting in silico experiments, which are cheaper and less tedious than real laboratory experiments. Results This paper aims at developing an extensible and computationally efficient framework for in silico modeling of tumor growth in a 3-dimensional, inhomogeneous and time-varying chemical environment. The resulting model consists of a set of mathematically derived and algorithmically defined operators, each one addressing the effects of a particular biological mechanism on the state of the system. These operators may be extended or re-adjusted, in case a different set of starting assumptions or a different simulation scenario needs to be considered. Conclusion In silico modeling provides an alternative means for testing hypotheses and simulating scenarios for which exact biological knowledge remains elusive. However, finer tuning of pertinent methods presupposes qualitative and quantitative enrichment of available biological evidence. Validation in a strict sense would further require comprehensive, case-specific simulations and detailed comparisons with biomedical observations.
Collapse
Affiliation(s)
- Markos Antonopoulos
- Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece.
| | - Dimitra Dionysiou
- Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| | - Georgios Stamatakos
- Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| | - Nikolaos Uzunoglu
- Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| |
Collapse
|
10
|
Lai X, Geier OM, Fleischer T, Garred Ø, Borgen E, Funke SW, Kumar S, Rognes ME, Seierstad T, Børresen-Dale AL, Kristensen VN, Engebraaten O, Köhn-Luque A, Frigessi A. Toward Personalized Computer Simulation of Breast Cancer Treatment: A Multiscale Pharmacokinetic and Pharmacodynamic Model Informed by Multitype Patient Data. Cancer Res 2019; 79:4293-4304. [PMID: 31118201 PMCID: PMC7617071 DOI: 10.1158/0008-5472.can-18-1804] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/13/2019] [Accepted: 05/17/2019] [Indexed: 11/16/2022]
Abstract
The usefulness of mechanistic models to disentangle complex multiscale cancer processes, such as treatment response, has been widely acknowledged. However, a major barrier for multiscale models to predict treatment outcomes in individual patients lies in their initialization and parametrization, which needs to reflect individual cancer characteristics accurately. In this study, we use multitype measurements acquired routinely on a single breast tumor, including histopathology, MRI, and molecular profiling, to personalize parts of a complex multiscale model of breast cancer treated with chemotherapeutic and antiangiogenic agents. The model accounts for drug pharmacokinetics and pharmacodynamics. We developed an open-source computer program that simulates cross-sections of tumors under 12-week therapy regimens and used it to individually reproduce and elucidate treatment outcomes of 4 patients. Two of the tumors did not respond to therapy, and model simulations were used to suggest alternative regimens with improved outcomes dependent on the tumor's individual characteristics. It was determined that more frequent and lower doses of chemotherapy reduce tumor burden in a low proliferative tumor while lower doses of antiangiogenic agents improve drug penetration in a poorly perfused tumor. Furthermore, using this model, we were able to correctly predict the outcome in another patient after 12 weeks of treatment. In summary, our model bridges multitype clinical data to shed light on individual treatment outcomes. SIGNIFICANCE: Mathematical modeling is used to validate possible mechanisms of tumor growth, resistance, and treatment outcome.
Collapse
Affiliation(s)
- Xiaoran Lai
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Oliver M Geier
- Department of Diagnostic Physics, Clinic of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Thomas Fleischer
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Simon W Funke
- Center for Biomedical Computing, Simula Research Laboratory, Lysaker, Norway
| | - Surendra Kumar
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Marie E Rognes
- Center for Biomedical Computing, Simula Research Laboratory, Lysaker, Norway
| | - Therese Seierstad
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Vessela N Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Molecular Biology and Laboratory Science (EpiGen), Division of Medicine, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Olav Engebraaten
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, University of Oslo, Oslo, Norway
| | - Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Arnoldo Frigessi
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
11
|
Forster JC, Marcu LG, Bezak E. Approaches to combat hypoxia in cancer therapy and the potential for in silico models in their evaluation. Phys Med 2019; 64:145-156. [PMID: 31515013 DOI: 10.1016/j.ejmp.2019.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/17/2019] [Accepted: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
AIM The negative impact of tumour hypoxia on cancer treatment outcome has been long-known, yet there has been little success combating it. This paper investigates the potential role of in silico modelling to help test emerging hypoxia-targeting treatments in cancer therapy. METHODS A Medline search was undertaken on the current landscape of in silico models that simulate cancer therapy and evaluate their ability to test hypoxia-targeting treatments. Techniques and treatments to combat tumour hypoxia and their current challenges are also presented. RESULTS Hypoxia-targeting treatments include tumour reoxygenation, hypoxic cell radiosensitization with nitroimidazoles, hypoxia-activated prodrugs and molecular targeting. Their main challenges are toxicity and not achieving adequate delivery to hypoxic regions of the tumour. There is promising research toward combining two or more of these techniques. Different types of in silico therapy models have been developed ranging from temporal to spatial and from stochastic to deterministic models. Numerous models have compared the effectiveness of different radiotherapy fractionation schedules for controlling hypoxic tumours. Similarly, models could help identify and optimize new treatments for overcoming hypoxia that utilize novel hypoxia-targeting technology. CONCLUSION Current therapy models should attempt to incorporate more sophisticated modelling of tumour angiogenesis/vasculature and vessel perfusion in order to become more useful for testing hypoxia-targeting treatments, which typically rely upon the tumour vasculature for delivery of additional oxygen, (pro)drugs and nanoparticles.
Collapse
Affiliation(s)
- Jake C Forster
- SA Medical Imaging, Department of Nuclear Medicine, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; Department of Physics, University of Adelaide, North Terrace, Adelaide SA 5005, Australia
| | - Loredana G Marcu
- Faculty of Science, University of Oradea, Oradea 410087, Romania; Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA 5001, Australia.
| | - Eva Bezak
- Department of Physics, University of Adelaide, North Terrace, Adelaide SA 5005, Australia; Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide SA 5001, Australia
| |
Collapse
|
12
|
Kyroudis CA, Dionysiou DD, Kolokotroni EA, Stamatakos GS. Studying the regression profiles of cervical tumours during radiotherapy treatment using a patient-specific multiscale model. Sci Rep 2019; 9:1081. [PMID: 30705291 PMCID: PMC6355788 DOI: 10.1038/s41598-018-37155-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/03/2018] [Indexed: 12/24/2022] Open
Abstract
Apart from offering insight into the biomechanisms involved in cancer, many recent mathematical modeling efforts aspire to the ultimate goal of clinical translation, wherein models are designed to be used in the future as clinical decision support systems in the patient-individualized context. Most significant challenges are the integration of multiscale biodata and the patient-specific model parameterization. A central aim of this study was the design of a clinically-relevant parameterization methodology for a patient-specific computational model of cervical cancer response to radiotherapy treatment with concomitant cisplatin, built around a tumour features-based search of the parameter space. Additionally, a methodological framework for the predictive use of the model was designed, including a scoring method to quantitatively reflect the similarity and bilateral predictive ability of any two tumours in terms of their regression profile. The methodology was applied to the datasets of eight patients. Tumour scenarios in accordance with the available longitudinal data have been determined. Predictive investigations identified three patient cases, anyone of which can be used to predict the volumetric evolution throughout therapy of the tumours of the other two with very good results. Our observations show that the presented approach is promising in quantifiably differentiating tumours with distinct regression profiles.
Collapse
Affiliation(s)
- Christos A Kyroudis
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Dimitra D Dionysiou
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece.
| | - Eleni A Kolokotroni
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Georgios S Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| |
Collapse
|
13
|
Ouzounoglou E, Kolokotroni E, Stanulla M, Stamatakos GS. A study on the predictability of acute lymphoblastic leukaemia response to treatment using a hybrid oncosimulator. Interface Focus 2018; 8:20160163. [PMID: 29285342 PMCID: PMC5740218 DOI: 10.1098/rsfs.2016.0163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Efficient use of Virtual Physiological Human (VPH)-type models for personalized treatment response prediction purposes requires a precise model parameterization. In the case where the available personalized data are not sufficient to fully determine the parameter values, an appropriate prediction task may be followed. This study, a hybrid combination of computational optimization and machine learning methods with an already developed mechanistic model called the acute lymphoblastic leukaemia (ALL) Oncosimulator which simulates ALL progression and treatment response is presented. These methods are used in order for the parameters of the model to be estimated for retrospective cases and to be predicted for prospective ones. The parameter value prediction is based on a regression model trained on retrospective cases. The proposed Hybrid ALL Oncosimulator system has been evaluated when predicting the pre-phase treatment outcome in ALL. This has been correctly achieved for a significant percentage of patient cases tested (approx. 70% of patients). Moreover, the system is capable of denying the classification of cases for which the results are not trustworthy enough. In that case, potentially misleading predictions for a number of patients are avoided, while the classification accuracy for the remaining patient cases further increases. The results obtained are particularly encouraging regarding the soundness of the proposed methodologies and their relevance to the process of achieving clinical applicability of the proposed Hybrid ALL Oncosimulator system and VPH models in general.
Collapse
Affiliation(s)
- Eleftherios Ouzounoglou
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| | - Eleni Kolokotroni
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| | - Martin Stanulla
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Georgios S Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| |
Collapse
|
14
|
Caballero D, Kaushik S, Correlo V, Oliveira J, Reis R, Kundu S. Organ-on-chip models of cancer metastasis for future personalized medicine: From chip to the patient. Biomaterials 2017; 149:98-115. [DOI: 10.1016/j.biomaterials.2017.10.005] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/15/2017] [Accepted: 10/02/2017] [Indexed: 02/09/2023]
|