1
|
Wertheim KY, Chisholm R, Richmond P, Walker D. Multicellular model of neuroblastoma proposes unconventional therapy based on multiple roles of p53. PLoS Comput Biol 2024; 20:e1012648. [PMID: 39715281 DOI: 10.1371/journal.pcbi.1012648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Neuroblastoma is the most common extra-cranial solid tumour in children. Over half of all high-risk cases are expected to succumb to the disease even after chemotherapy, surgery, and immunotherapy. Although the importance of MYCN amplification in this disease is indisputable, the mechanistic details remain enigmatic. Here, we present a multicellular model of neuroblastoma comprising a continuous automaton, discrete cell agents, and a centre-based mechanical model, as well as the simulation results we obtained with it. The continuous automaton represents the tumour microenvironment as a grid-like structure, where each voxel is associated with continuous variables such as the oxygen level therein. Each discrete cell agent is defined by several attributes, including its cell cycle position, mutations, gene expression pattern, and more with behaviours such as cell cycling and cell death being stochastically dependent on these attributes. The centre-based mechanical model represents the properties of these agents as physical objects, describing how they repel each other as soft spheres. By implementing a stochastic simulation algorithm on modern GPUs, we simulated the dynamics of over one million neuroblastoma cells over a period of months. Specifically, we set up 1200 heterogeneous tumours and tracked the MYCN-amplified clone's dynamics in each, revealed the conditions that favour its growth, and tested its responses to 5000 drug combinations. Our results are in agreement with those reported in the literature and add new insights into how the MYCN-amplified clone's reproductive advantage in a tumour, its gene expression profile, the tumour's other clones (with different mutations), and the tumour's microenvironment are inter-related. Based on the results, we formulated a hypothesis, which argues that there are two distinct populations of neuroblastoma cells in the tumour; the p53 protein is pro-survival in one and pro-apoptosis in the other. It follows that alternating between inhibiting MDM2 to restore p53 activity and inhibiting ARF to attenuate p53 activity is a promising, if unorthodox, therapeutic strategy. The multicellular model has the advantages of modularity, high resolution, and scalability, making it a potential foundation for creating digital twins of neuroblastoma patients.
Collapse
Affiliation(s)
- Kenneth Y Wertheim
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
- Centre of Excellence for Data Science, Artificial Intelligence, and Modelling, University of Hull, Kingston upon Hull, United Kingdom
- School of Computer Science, University of Hull, Kingston upon Hull, United Kingdom
| | - Robert Chisholm
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Paul Richmond
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Dawn Walker
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- Department of Computer Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
2
|
Wang H, Chen X, Li T, Xie M, Qin J, Zhang L, Ding H, He L. Identification of an Ultra-High-Risk Subgroup of Neuroblastoma Patients within the High-Risk Cohort Using a Computed Tomography-Based Radiomics Approach. Acad Radiol 2024; 31:1655-1665. [PMID: 37714717 DOI: 10.1016/j.acra.2023.08.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/17/2023]
Abstract
RATIONALE AND OBJECTIVES To identify ultra-high-risk (UHR) neuroblastoma patients who experienced disease-related mortality within 18 months of diagnosis within the high-risk cohort using computed tomography (CT)-based radiomics analysis. MATERIALS AND METHODS A retrospective analysis was conducted on 105 high-risk neuroblastoma patients, divided into a training set (n = 74) and a test set (n = 31). Radiomics features were extracted and selected from arterial phase CT images, and an optimal radiomics signature was established using the support vector machine algorithm. Evaluation metrics, including area under the curve (AUC) and 95% confidence interval (CI), were calculated. Furthermore, the fit and clinical benefit of the signature, along with its correlation with overall survival (OS), were analyzed. RESULTS The optimal radiomics signature comprised 11 features. In the training set, AUC and accuracy were 0.911 (95% CI: 0.840-0.982) and 0.892, respectively. In the test set, AUC and accuracy were 0.828 (95% CI: 0.669-0.987) and 0.839, respectively. There was no significant difference between predicted probability and actual probability, and the signature demonstrated net benefit. The concordance index of this signature for predicting OS was 0.743 (95% CI: 0.672-0.814) in the training set and 0.688 (95% CI: 0.566-0.810) in the test set. Moreover, the signature achieved AUC values of 0.832, 0.863, and 0.721 for 1-year, 2-year, and 3-year OS in the training set, and 0.870, 0.836, and 0.638 in the test set for the respective time periods. CONCLUSION The utilization of CT-based radiomics signature to identify an UHR subgroup of neuroblastoma patients within the high-risk cohort can help aid in predicting early disease progression.
Collapse
Affiliation(s)
- Haoru Wang
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China
| | - Xin Chen
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China
| | - Ting Li
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China
| | - Mingye Xie
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China
| | - Jinjie Qin
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China
| | - Li Zhang
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China
| | - Hao Ding
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China
| | - Ling He
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014, China.
| |
Collapse
|
3
|
Metzcar J, Jutzeler CR, Macklin P, Köhn-Luque A, Brüningk SC. A review of mechanistic learning in mathematical oncology. Front Immunol 2024; 15:1363144. [PMID: 38533513 PMCID: PMC10963621 DOI: 10.3389/fimmu.2024.1363144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024] Open
Abstract
Mechanistic learning refers to the synergistic combination of mechanistic mathematical modeling and data-driven machine or deep learning. This emerging field finds increasing applications in (mathematical) oncology. This review aims to capture the current state of the field and provides a perspective on how mechanistic learning may progress in the oncology domain. We highlight the synergistic potential of mechanistic learning and point out similarities and differences between purely data-driven and mechanistic approaches concerning model complexity, data requirements, outputs generated, and interpretability of the algorithms and their results. Four categories of mechanistic learning (sequential, parallel, extrinsic, intrinsic) of mechanistic learning are presented with specific examples. We discuss a range of techniques including physics-informed neural networks, surrogate model learning, and digital twins. Example applications address complex problems predominantly from the domain of oncology research such as longitudinal tumor response predictions or time-to-event modeling. As the field of mechanistic learning advances, we aim for this review and proposed categorization framework to foster additional collaboration between the data- and knowledge-driven modeling fields. Further collaboration will help address difficult issues in oncology such as limited data availability, requirements of model transparency, and complex input data which are embraced in a mechanistic learning framework.
Collapse
Affiliation(s)
- John Metzcar
- Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Bloomington, IN, United States
- Informatics, Luddy School of Informatics, Computing, and Engineering, Bloomington, IN, United States
| | - Catherine R. Jutzeler
- Department of Health Sciences and Technology (D-HEST), Eidgenössische Technische Hochschule Zürich (ETH), Zürich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Paul Macklin
- Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Bloomington, IN, United States
| | - Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, Oslo, Norway
| | - Sarah C. Brüningk
- Department of Health Sciences and Technology (D-HEST), Eidgenössische Technische Hochschule Zürich (ETH), Zürich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| |
Collapse
|
4
|
Benzekry S, Schlicke P, Mogenet A, Greillier L, Tomasini P, Simon E. Computational markers for personalized prediction of outcomes in non-small cell lung cancer patients with brain metastases. Clin Exp Metastasis 2024; 41:55-68. [PMID: 38117432 DOI: 10.1007/s10585-023-10245-3] [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: 09/18/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2023]
Abstract
Intracranial progression after curative treatment of early-stage non-small cell lung cancer (NSCLC) occurs from 10 to 50% and is difficult to manage, given the heterogeneity of clinical presentations and the variability of treatments available. The objective of this study was to develop a mechanistic model of intracranial progression to predict survival following a first brain metastasis (BM) event occurring at a time [Formula: see text]. Data included early-stage NSCLC patients treated with a curative intent who had a BM as the first and single relapse site (N = 31). We propose a mechanistic mathematical model able to derive computational markers from primary tumor and BM data at [Formula: see text] and estimate the amount and sizes of (visible and invisible) BMs, as well as their future behavior. These two key computational markers are [Formula: see text], the proliferation rate of a single tumor cell; and [Formula: see text], the per day, per cell, probability to metastasize. The predictive value of these individual computational biomarkers was evaluated. The model was able to correctly describe the number and size of metastases at [Formula: see text] for 20 patients. Parameters [Formula: see text] and [Formula: see text] were significantly associated with overall survival (OS) (HR 1.65 (1.07-2.53) p = 0.0029 and HR 1.95 (1.31-2.91) p = 0.0109, respectively). Adding the computational markers to the clinical ones significantly improved the predictive value of OS (c-index increased from 0.585 (95% CI 0.569-0.602) to 0.713 (95% CI 0.700-0.726), p < 0.0001). We demonstrated that our model was applicable to brain oligoprogressive patients in NSCLC and that the resulting computational markers had predictive potential. This may help lung cancer physicians to guide and personalize the management of NSCLC patients with intracranial oligoprogression.
Collapse
Affiliation(s)
- Sébastien Benzekry
- COMPutational Pharmacology and Clinical Oncology Department, Inria Sophia Antipolis - Méditerranée, Faculté de Pharmacie, Cancer Research Center of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, 27 Boulevard Jean Moulin, 13005, Marseille, France.
| | - Pirmin Schlicke
- Department of Mathematics, TUM School of Computation, Information and Technology, Technical University of Munich, Garching (Munich), Germany
| | - Alice Mogenet
- Multidisciplinary Oncology and Therapeutic Innovations Department, Assistance Publique - Hôpitaux de Marseille, Aix Marseille University, Marseille, France
| | - Laurent Greillier
- Multidisciplinary Oncology and Therapeutic Innovations Department, Assistance Publique - Hôpitaux de Marseille, Aix Marseille University, Marseille, France
- Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France
| | - Pascale Tomasini
- Multidisciplinary Oncology and Therapeutic Innovations Department, Assistance Publique - Hôpitaux de Marseille, Aix Marseille University, Marseille, France
- Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France
| | - Eléonore Simon
- Multidisciplinary Oncology and Therapeutic Innovations Department, Assistance Publique - Hôpitaux de Marseille, Aix Marseille University, Marseille, France
| |
Collapse
|
5
|
Borau C, Wertheim KY, Hervas-Raluy S, Sainz-DeMena D, Walker D, Chisholm R, Richmond P, Varella V, Viceconti M, Montero A, Gregori-Puigjané E, Mestres J, Kasztelnik M, García-Aznar JM. A multiscale orchestrated computational framework to reveal emergent phenomena in neuroblastoma. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 241:107742. [PMID: 37572512 DOI: 10.1016/j.cmpb.2023.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/19/2023] [Accepted: 07/31/2023] [Indexed: 08/14/2023]
Abstract
Neuroblastoma is a complex and aggressive type of cancer that affects children. Current treatments involve a combination of surgery, chemotherapy, radiotherapy, and stem cell transplantation. However, treatment outcomes vary due to the heterogeneous nature of the disease. Computational models have been used to analyse data, simulate biological processes, and predict disease progression and treatment outcomes. While continuum cancer models capture the overall behaviour of tumours, and agent-based models represent the complex behaviour of individual cells, multiscale models represent interactions at different organisational levels, providing a more comprehensive understanding of the system. In 2018, the PRIMAGE consortium was formed to build a cloud-based decision support system for neuroblastoma, including a multi-scale model for patient-specific simulations of disease progression. In this work we have developed this multi-scale model that includes data such as patient's tumour geometry, cellularity, vascularization, genetics and type of chemotherapy treatment, and integrated it into an online platform that runs the simulations on a high-performance computation cluster using Onedata and Kubernetes technologies. This infrastructure will allow clinicians to optimise treatment regimens and reduce the number of costly and time-consuming clinical trials. This manuscript outlines the challenging framework's model architecture, data workflow, hypothesis, and resources employed in its development.
Collapse
Affiliation(s)
- C Borau
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Mechanical Engineering Department, University of Zaragoza, Zaragoza, Spain.
| | - K Y Wertheim
- Department of Computer Science and InsigneoInstitute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom; Centre of Excellence for Data Science, Artificial Intelligence and Modelling and School of Computer Science, University of Hull, Kingston upon Hull, United Kingdom
| | - S Hervas-Raluy
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Mechanical Engineering Department, University of Zaragoza, Zaragoza, Spain
| | - D Sainz-DeMena
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Mechanical Engineering Department, University of Zaragoza, Zaragoza, Spain
| | - D Walker
- Department of Computer Science and InsigneoInstitute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - R Chisholm
- Department of Computer Science and InsigneoInstitute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - P Richmond
- Department of Computer Science and InsigneoInstitute for In Silico Medicine, University of Sheffield, Sheffield, United Kingdom
| | - V Varella
- Department of Industrial Engineering, Alma Mater Studiorum - University of Bologna, Bologna, Italy; Medical Technology Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - M Viceconti
- Department of Industrial Engineering, Alma Mater Studiorum - University of Bologna, Bologna, Italy; Medical Technology Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - A Montero
- Chemotargets SL, Baldiri Reixac 4, Parc Cientific de Barcelona (PCB), Barcelona, Spain
| | - E Gregori-Puigjané
- Chemotargets SL, Baldiri Reixac 4, Parc Cientific de Barcelona (PCB), Barcelona, Spain
| | - J Mestres
- Chemotargets SL, Baldiri Reixac 4, Parc Cientific de Barcelona (PCB), Barcelona, Spain
| | - M Kasztelnik
- ACC Cyfronet, AGH University of Science and Technology, Kraków, Poland
| | - J M García-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Mechanical Engineering Department, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
6
|
Wagner A, Schlicke P, Fritz M, Kuttler C, Oden JT, Schumann C, Wohlmuth B. A phase-field model for non-small cell lung cancer under the effects of immunotherapy. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:18670-18694. [PMID: 38052574 DOI: 10.3934/mbe.2023828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Formulating mathematical models that estimate tumor growth under therapy is vital for improving patient-specific treatment plans. In this context, we present our recent work on simulating non-small-scale cell lung cancer (NSCLC) in a simple, deterministic setting for two different patients receiving an immunotherapeutic treatment. At its core, our model consists of a Cahn-Hilliard-based phase-field model describing the evolution of proliferative and necrotic tumor cells. These are coupled to a simplified nutrient model that drives the growth of the proliferative cells and their decay into necrotic cells. The applied immunotherapy decreases the proliferative cell concentration. Here, we model the immunotherapeutic agent concentration in the entire lung over time by an ordinary differential equation (ODE). Finally, reaction terms provide a coupling between all these equations. By assuming spherical, symmetric tumor growth and constant nutrient inflow, we simplify this full 3D cancer simulation model to a reduced 1D model. We can then resort to patient data gathered from computed tomography (CT) scans over several years to calibrate our model. Our model covers the case in which the immunotherapy is successful and limits the tumor size, as well as the case predicting a sudden relapse, leading to exponential tumor growth. Finally, we move from the reduced model back to the full 3D cancer simulation in the lung tissue. Thereby, we demonstrate the predictive benefits that a more detailed patient-specific simulation including spatial information as a possible generalization within our framework could yield in the future.
Collapse
Affiliation(s)
- Andreas Wagner
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| | - Pirmin Schlicke
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| | - Marvin Fritz
- Computational Methods for PDEs, Johann Radon Institute for Computational and Applied Mathematics, Linz, Upper Austria, Austria
| | - Christina Kuttler
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| | - J Tinsley Oden
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Christian Schumann
- Clinic of Pneumology, Thoracic Oncology, Sleep and Respiratory Critical Care, Klinikverbund Allgäu, Kempten, Bavaria, Germany
| | - Barbara Wohlmuth
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| |
Collapse
|
7
|
Italia M, Wertheim KY, Taschner-Mandl S, Walker D, Dercole F. Mathematical Model of Clonal Evolution Proposes a Personalised Multi-Modal Therapy for High-Risk Neuroblastoma. Cancers (Basel) 2023; 15:cancers15071986. [PMID: 37046647 PMCID: PMC10093626 DOI: 10.3390/cancers15071986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Neuroblastoma is the most common extra-cranial solid tumour in children. Despite multi-modal therapy, over half of the high-risk patients will succumb. One contributing factor is the one-size-fits-all nature of multi-modal therapy. For example, during the first step (induction chemotherapy), the standard regimen (rapid COJEC) administers fixed doses of chemotherapeutic agents in eight two-week cycles. Perhaps because of differences in resistance, this standard regimen results in highly heterogeneous outcomes in different tumours. In this study, we formulated a mathematical model comprising ordinary differential equations. The equations describe the clonal evolution within a neuroblastoma tumour being treated with vincristine and cyclophosphamide, which are used in the rapid COJEC regimen, including genetically conferred and phenotypic drug resistance. The equations also describe the agents’ pharmacokinetics. We devised an optimisation algorithm to find the best chemotherapy schedules for tumours with different pre-treatment clonal compositions. The optimised chemotherapy schedules exploit the cytotoxic difference between the two drugs and intra-tumoural clonal competition to shrink the tumours as much as possible during induction chemotherapy and before surgical removal. They indicate that induction chemotherapy can be improved by finding and using personalised schedules. More broadly, we propose that the overall multi-modal therapy can be enhanced by employing targeted therapies against the mutations and oncogenic pathways enriched and activated by the chemotherapeutic agents. To translate the proposed personalised multi-modal therapy into clinical use, patient-specific model calibration and treatment optimisation are necessary. This entails a decision support system informed by emerging medical technologies such as multi-region sequencing and liquid biopsies. The results and tools presented in this paper could be the foundation of this decision support system.
Collapse
Affiliation(s)
- Matteo Italia
- Department of Electronic, Information, and Bioengineering, Politecnico di Milano, 20133 Milano, Italy
- Correspondence:
| | - Kenneth Y. Wertheim
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield S10 2TN, UK
- Department of Computer Science, University of Sheffield, Sheffield S10 2TN, UK
- Centre of Excellence for Data Science, Artificial Intelligence, and Modelling, University of Hull, Kingston upon Hull HU6 7RX, UK
- School of Computer Science, University of Hull, Kingston upon Hull HU6 7RX, UK
| | | | - Dawn Walker
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield S10 2TN, UK
- Department of Computer Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Fabio Dercole
- Department of Electronic, Information, and Bioengineering, Politecnico di Milano, 20133 Milano, Italy
| |
Collapse
|
8
|
Álvarez-Arenas A, Souleyreau W, Emanuelli A, Cooley LS, Bernhard JC, Bikfalvi A, Benzekry S. Practical identifiability analysis of a mechanistic model for the time to distant metastatic relapse and its application to renal cell carcinoma. PLoS Comput Biol 2022; 18:e1010444. [PMID: 36007057 PMCID: PMC9451098 DOI: 10.1371/journal.pcbi.1010444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/07/2022] [Accepted: 07/27/2022] [Indexed: 12/02/2022] Open
Abstract
Distant metastasis-free survival (DMFS) curves are widely used in oncology. They are classically analyzed using the Kaplan-Meier estimator or agnostic statistical models from survival analysis. Here we report on a method to extract more information from DMFS curves using a mathematical model of primary tumor growth and metastatic dissemination. The model depends on two parameters, α and μ, respectively quantifying tumor growth and dissemination. We assumed these to be lognormally distributed in a patient population. We propose a method for identification of the parameters of these distributions based on least-squares minimization between the data and the simulated survival curve. We studied the practical identifiability of these parameters and found that including the percentage of patients with metastasis at diagnosis was critical to ensure robust estimation. We also studied the impact and identifiability of covariates and their coefficients in α and μ, either categorical or continuous, including various functional forms for the latter (threshold, linear or a combination of both). We found that both the functional form and the coefficients could be determined from DMFS curves. We then applied our model to a clinical dataset of metastatic relapse from kidney cancer with individual data of 105 patients. We show that the model was able to describe the data and illustrate our method to disentangle the impact of three covariates on DMFS: a categorical one (Führman grade) and two continuous ones (gene expressions of the macrophage mannose receptor 1 (MMR) and the G Protein-Coupled Receptor Class C Group 5 Member A (GPRC5a) gene). We found that all had an influence in metastasis dissemination (μ), but not on growth (α). Understanding biological mechanisms leading to metastasis development is a major challenge in order to prevent distant relapse of cancer. Classical methods to study associations of biomarkers with subsequent metastatic relapse rely on the analysis of metastasis free survival curves by means of statistical models such as proportional hazards Cox regression. These models act as black boxes and don’t provide detailed information about the specific mechanism involved. In our study, we propose to use a method based on mechanistic modeling of the metastatic development, that is, a mathematical model that simulates the biological process. The main challenge for these models is to implement the right level of complexity, because if too many parameters are included, these cannot be precisely identified from the data. We reduced the metastatic process to two main aspects: growth and dissemination. We then proposed a theoretical study of the identifiability of the two associated parameters from metastasis-free survival curves. Eventually, we applied our method to a clinical dataset in kidney cancer and illustrated how we could gain biological insights about the role of some diagnosis markers.
Collapse
Affiliation(s)
| | | | - Andrea Emanuelli
- University of Bordeaux, LAMC, Pessac, France
- Inserm U1029, Pessac, France
| | - Lindsay S. Cooley
- University of Bordeaux, LAMC, Pessac, France
- Inserm U1029, Pessac, France
| | | | - Andreas Bikfalvi
- University of Bordeaux, LAMC, Pessac, France
- Inserm U1029, Pessac, France
| | - Sebastien Benzekry
- COMPO, COMPutational pharmacology and clinical Oncology, Centre Inria Sophia Antipolis - Méditerranée, Centre de Recherches en Cancérologie de Marseille, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix-Marseille University
- * E-mail:
| |
Collapse
|
9
|
Shameer K, Zhang Y, Prokop A, Nampally S, N IKA, Weatherall J, Iacona RB, Khan FM. OSPred Tool: A Digital Health Aid for Rapid Predictive Analysis of Correlations Between Early End Points and Overall Survival in Non-Small-Cell Lung Cancer Clinical Trials. JCO Clin Cancer Inform 2022; 6:e2100173. [PMID: 35467964 PMCID: PMC9067362 DOI: 10.1200/cci.21.00173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Overall survival (OS) is the gold standard end point for establishing clinical benefits in phase III oncology trials. However, these trials are associated with low success rates, largely driven by failure to meet the primary end point. Surrogate end points such as progression-free survival (PFS) are increasingly being used as indicators of biologic drug activity and to inform early go/no-go decisions in oncology drug development. We developed OSPred, a digital health aid that combines actual clinical data and machine intelligence approaches to visualize correlation trends between early (PFS-based) and late (OS) end points and provide support for shared decision making in the drug development pipeline.
Collapse
Affiliation(s)
- Khader Shameer
- Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Youyi Zhang
- Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Andrzej Prokop
- Oncology Biometrics, Oncology R&D, AstraZeneca, Warsaw, Poland
| | - Sreenath Nampally
- Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Imran Khan A N
- Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Macclesfield, United Kingdom
| | - Jim Weatherall
- Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Macclesfield, United Kingdom
| | | | - Faisal M Khan
- Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| |
Collapse
|
10
|
Zhang J, Liu W, Ji P, Zhang Y. Silencing of long chain noncoding RNA paternally expressed gene (PEG10) inhibits the progression of neuroblastoma by regulating microRNA-449a (miR-449a)/ribosomal protein S2 (RPS2) axis. Bioengineered 2022; 13:6309-6322. [PMID: 35212607 PMCID: PMC8973610 DOI: 10.1080/21655979.2022.2042999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
To investigate the mechanism of paternally expressed gene (PEG10) in regulating neuroblastoma (NB) progression. PEG10 expression was detected using quantitative real-time reverse transcription polymerase-chain reaction (qRT-PCR). The interaction of miR-449a and PEG10 or ribosomal protein S2 (RPS2) was employed by starBase, and then proved through RIP and dual-luciferase reporter assays. The NB cell viability, proliferation, invasion, and migration were evaluated by Cell Counting Kit-8 (CCK-8), colony formation, and Transwell assay. The mRNA and protein levels were determined by qRT-PCR and Western blotting, respectively. The levels of PEG10 and RPS2 were remarkably increased in NB tissues and cells, nevertheless the expression of miR-449a was conspicuously declined in NB tissues and cells. Silencing of PEG10 inhibited proliferation, migration, and invasion in SK-N-BE (2) cells, while overexpression of PEG10 promoted proliferation, migration, and invasion in SH-SY5Y cells. We affirmed that PEG10 interacted with miR-449a, and miR-449a could target the 3'UTR of RPS2 and negatively regulate its expression in NB cells. The upregulation of miR-449a inhibited proliferation, migration, and invasion in SK-N-BE (2) cells, while downregulation of miR-449a promoted proliferation, migration, and invasion in SH-SY5Y cells. Moreover, miR-449a overexpression weaken the function of PEG10-mediated on promoting proliferation, migration, and invasion in SH-SY5Y cells, while RPS2 overexpression rescued the effects of miR-449a-mediated on inhibiting those behaviors of SH-SY5Y cells. In conclusion, Silencing of PEG10 could inhibit proliferation, migration, and invasion via the miR-449a/RPS2 axis in NB cells.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Geriatrics, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, P.R. China
| | - Wei Liu
- Department of Health Management, Qingdao Eighth People's Hospital, Qingdao, Shandong, P.R. China
| | - Ping Ji
- Department of Ophthalmology, Qingdao Eighth People's Hospital, Qingdao, Shandong, P.R. China
| | - Yan Zhang
- The Third Department of Internal Medicine, Qingdao Women and Children's Hospital, Qingdao University, Qingdao, Shandong, P.R. China
| |
Collapse
|