1
|
Tavakoli N, Fong EJ, Coleman A, Huang YK, Bigger M, Doche ME, Kim S, Lenz HJ, Graham NA, Macklin P, Finley SD, Mumenthaler SM. Merging Metabolic Modeling and Imaging for Screening Therapeutic Targets in Colorectal Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595756. [PMID: 38826317 PMCID: PMC11142224 DOI: 10.1101/2024.05.24.595756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in metabolic reprogramming and are well-established contributors to drug resistance in colorectal cancer (CRC). To exploit this metabolic crosstalk, we integrated a systems biology approach that identified key metabolic targets in a data-driven method and validated them experimentally. This process involved a novel machine learning-based method to computationally screen, in a high-throughput manner, the effects of enzyme perturbations predicted by a computational model of CRC metabolism. This approach reveals the network-wide effects of metabolic perturbations. Our results highlighted hexokinase (HK) as a crucial target, which subsequently became our focus for experimental validation using patient-derived tumor organoids (PDTOs). Through metabolic imaging and viability assays, we found that PDTOs cultured in CAF-conditioned media exhibited increased sensitivity to HK inhibition, confirming the model predictions. Our approach emphasizes the critical role of integrating computational and experimental techniques in exploring and exploiting CRC-CAF crosstalk.
Collapse
|
2
|
Del Pino Herrera A, Ferrall-Fairbanks MC. A war on many fronts: cross disciplinary approaches for novel cancer treatment strategies. Front Genet 2024; 15:1383676. [PMID: 38873108 PMCID: PMC11169904 DOI: 10.3389/fgene.2024.1383676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/26/2024] [Indexed: 06/15/2024] Open
Abstract
Cancer is a disease characterized by uncontrolled cellular growth where cancer cells take advantage of surrounding cellular populations to obtain resources and promote invasion. Carcinomas are the most common type of cancer accounting for almost 90% of cancer cases. One of the major subtypes of carcinomas are adenocarcinomas, which originate from glandular cells that line certain internal organs. Cancers such as breast, prostate, lung, pancreas, colon, esophageal, kidney are often adenocarcinomas. Current treatment strategies include surgery, chemotherapy, radiation, targeted therapy, and more recently immunotherapy. However, patients with adenocarcinomas often develop resistance or recur after the first line of treatment. Understanding how networks of tumor cells interact with each other and the tumor microenvironment is crucial to avoid recurrence, resistance, and high-dose therapy toxicities. In this review, we explore how mathematical modeling tools from different disciplines can aid in the development of effective and personalized cancer treatment strategies. Here, we describe how concepts from the disciplines of ecology and evolution, economics, and control engineering have been applied to mathematically model cancer dynamics and enhance treatment strategies.
Collapse
Affiliation(s)
- Adriana Del Pino Herrera
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Meghan C. Ferrall-Fairbanks
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
3
|
Dobreva A, Camacho ET, Miranda M. Mathematical model for glutathione dynamics in the retina. Sci Rep 2023; 13:10996. [PMID: 37419948 PMCID: PMC10328985 DOI: 10.1038/s41598-023-37938-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/29/2023] [Indexed: 07/09/2023] Open
Abstract
The retina is highly susceptible to the generation of toxic reactive oxygen species (ROS) that disrupt the normal operations of retinal cells. The glutathione (GSH) antioxidant system plays an important role in mitigating ROS. To perform its protective functions, GSH depends on nicotinamide adenine dinucleotide phosphate (NADPH) produced through the pentose phosphate pathway. This work develops the first mathematical model for the GSH antioxidant system in the outer retina, capturing the most essential components for formation of ROS, GSH production, its oxidation in detoxifying ROS, and subsequent reduction by NADPH. We calibrate and validate the model using experimental measurements, at different postnatal days up to PN28, from control mice and from the rd1 mouse model for the disease retinitis pigmentosa (RP). Global sensitivity analysis is then applied to examine the model behavior and identify the pathways with the greatest impact in control compared to RP conditions. The findings underscore the importance of GSH and NADPH production in dealing with oxidative stress during retinal development, especially after peak rod degeneration occurs in RP, leading to increased oxygen tension. This suggests that stimulation of GSH and NADPH synthesis could be a potential intervention strategy in degenerative mouse retinas with RP.
Collapse
Affiliation(s)
- Atanaska Dobreva
- Department of Mathematics, Augusta University, Augusta, GA, 30912, USA.
| | - Erika Tatiana Camacho
- University of Texas at San Antonio, San Antonio, TX, 78249, USA
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - María Miranda
- Department of Biomedical Sciences, Faculty of Health Sciences, Institute of Biomedical Sciences, Cardenal Herrera-CEU University, CEU Universities, 46115, Valencia, Spain
| |
Collapse
|
4
|
Yang J, Virostko J, Liu J, Jarrett AM, Hormuth DA, Yankeelov TE. Comparing mechanism-based and machine learning models for predicting the effects of glucose accessibility on tumor cell proliferation. Sci Rep 2023; 13:10387. [PMID: 37369672 DOI: 10.1038/s41598-023-37238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
Glucose plays a central role in tumor metabolism and development and is a target for novel therapeutics. To characterize the response of cancer cells to blockade of glucose uptake, we collected time-resolved microscopy data to track the growth of MDA-MB-231 breast cancer cells. We then developed a mechanism-based, mathematical model to predict how a glucose transporter (GLUT1) inhibitor (Cytochalasin B) influences the growth of the MDA-MB-231 cells by limiting access to glucose. The model includes a parameter describing dose dependent inhibition to quantify both the total glucose level in the system and the glucose level accessible to the tumor cells. Four common machine learning models were also used to predict tumor cell growth. Both the mechanism-based and machine learning models were trained and validated, and the prediction error was evaluated by the coefficient of determination (R2). The random forest model provided the highest accuracy predicting cell dynamics (R2 = 0.92), followed by the decision tree (R2 = 0.89), k-nearest-neighbor regression (R2 = 0.84), mechanism-based (R2 = 0.77), and linear regression model (R2 = 0.69). Thus, the mechanism-based model has a predictive capability comparable to machine learning models with the added benefit of elucidating biological mechanisms.
Collapse
Affiliation(s)
- Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keaton, BME Building, 1 University Station, C0800, Austin, TX, 78712, USA
| | - Jack Virostko
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Oncology, The University of Texas at Austin, Austin, TX, 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Junyan Liu
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keaton, BME Building, 1 University Station, C0800, Austin, TX, 78712, USA
| | - Angela M Jarrett
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, 78712, USA
| | - David A Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keaton, BME Building, 1 University Station, C0800, Austin, TX, 78712, USA.
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, 78712, USA.
- Department of Oncology, The University of Texas at Austin, Austin, TX, 78712, USA.
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA.
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, 78712, USA.
- Departments of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
5
|
Brummer AB, Xella A, Woodall R, Adhikarla V, Cho H, Gutova M, Brown CE, Rockne RC. Data driven model discovery and interpretation for CAR T-cell killing using sparse identification and latent variables. Front Immunol 2023; 14:1115536. [PMID: 37256133 PMCID: PMC10226275 DOI: 10.3389/fimmu.2023.1115536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/27/2023] [Indexed: 06/01/2023] Open
Abstract
In the development of cell-based cancer therapies, quantitative mathematical models of cellular interactions are instrumental in understanding treatment efficacy. Efforts to validate and interpret mathematical models of cancer cell growth and death hinge first on proposing a precise mathematical model, then analyzing experimental data in the context of the chosen model. In this work, we present the first application of the sparse identification of non-linear dynamics (SINDy) algorithm to a real biological system in order discover cell-cell interaction dynamics in in vitro experimental data, using chimeric antigen receptor (CAR) T-cells and patient-derived glioblastoma cells. By combining the techniques of latent variable analysis and SINDy, we infer key aspects of the interaction dynamics of CAR T-cell populations and cancer. Importantly, we show how the model terms can be interpreted biologically in relation to different CAR T-cell functional responses, single or double CAR T-cell-cancer cell binding models, and density-dependent growth dynamics in either of the CAR T-cell or cancer cell populations. We show how this data-driven model-discovery based approach provides unique insight into CAR T-cell dynamics when compared to an established model-first approach. These results demonstrate the potential for SINDy to improve the implementation and efficacy of CAR T-cell therapy in the clinic through an improved understanding of CAR T-cell dynamics.
Collapse
Affiliation(s)
- Alexander B. Brummer
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
- Department of Physics and Astronomy, College of Charleston, Charleston, SC, United States
| | - Agata Xella
- Department of Hemtaology and Hematopoietic Cell Translation and Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Ryan Woodall
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Vikram Adhikarla
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Heyrim Cho
- Department of Mathematics, University of California, Riverside, Riverside, CA, United States
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Christine E. Brown
- Department of Hemtaology and Hematopoietic Cell Translation and Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Russell C. Rockne
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
6
|
Miller HA, Miller DM, van Berkel VH, Frieboes HB. Evaluation of Lung Cancer Patient Response to First-Line Chemotherapy by Integration of Tumor Core Biopsy Metabolomics with Multiscale Modeling. Ann Biomed Eng 2023; 51:820-832. [PMID: 36224485 PMCID: PMC10023290 DOI: 10.1007/s10439-022-03096-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/02/2022] [Indexed: 11/28/2022]
Abstract
The standard of care for intermediate (Stage II) and advanced (Stages III and IV) non-small cell lung cancer (NSCLC) involves chemotherapy with taxane/platinum derivatives, with or without radiation. Ideally, patients would be screened a priori to allow non-responders to be initially treated with second-line therapies. This evaluation is non-trivial, however, since tumors behave as complex multiscale systems. To address this need, this study employs a multiscale modeling approach to evaluate first-line chemotherapy response of individual patient tumors based on metabolomic analysis of tumor core biopsies obtained during routine clinical evaluation. Model parameters were calculated for a patient cohort as a function of these metabolomic profiles, previously obtained from high-resolution 2DLC-MS/MS analysis. Evaluation metrics were defined to classify patients as Disease-Control (DC) [encompassing complete-response (CR), partial-response (PR), and stable-disease (SD)] and Progressive-Disease (PD) following first-line chemotherapy. Response was simulated for each patient and compared to actual response. The results show that patient classifications were significantly separated from each other, and also when grouped as DC vs. PD and as CR/PR vs. SD/PD, by fraction of initial tumor radius metric at 6 days post simulated bolus drug injection. This study shows that patient first-line chemotherapy response can in principle be evaluated from multiscale modeling integrated with tumor tissue metabolomic data, offering a first step towards individualized lung cancer treatment prognosis.
Collapse
Affiliation(s)
- Hunter A Miller
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Donald M Miller
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Victor H van Berkel
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, KY, USA
| | - Hermann B Frieboes
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA.
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Department of Bioengineering, University of Louisville, Lutz Hall 419, Louisville, KY, 40292, USA.
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
7
|
Gelbach PE, Zheng D, Fraser SE, White KL, Graham NA, Finley SD. Kinetic and data-driven modeling of pancreatic β-cell central carbon metabolism and insulin secretion. PLoS Comput Biol 2022; 18:e1010555. [PMID: 36251711 PMCID: PMC9612825 DOI: 10.1371/journal.pcbi.1010555] [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: 10/11/2021] [Revised: 10/27/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022] Open
Abstract
Pancreatic β-cells respond to increased extracellular glucose levels by initiating a metabolic shift. That change in metabolism is part of the process of glucose-stimulated insulin secretion and is of particular interest in the context of diabetes. However, we do not fully understand how the coordinated changes in metabolic pathways and metabolite products influence insulin secretion. In this work, we apply systems biology approaches to develop a detailed kinetic model of the intracellular central carbon metabolic pathways in pancreatic β-cells upon stimulation with high levels of glucose. The model is calibrated to published metabolomics datasets for the INS1 823/13 cell line, accurately capturing the measured metabolite fold-changes. We first employed the calibrated mechanistic model to estimate the stimulated cell's fluxome. We then used the predicted network fluxes in a data-driven approach to build a partial least squares regression model. By developing the combined kinetic and data-driven modeling framework, we gain insights into the link between β-cell metabolism and glucose-stimulated insulin secretion. The combined modeling framework was used to predict the effects of common anti-diabetic pharmacological interventions on metabolite levels, flux through the metabolic network, and insulin secretion. Our simulations reveal targets that can be modulated to enhance insulin secretion. The model is a promising tool to contextualize and extend the usefulness of metabolomics data and to predict dynamics and metabolite levels that are difficult to measure in vitro. In addition, the modeling framework can be applied to identify, explain, and assess novel and clinically-relevant interventions that may be particularly valuable in diabetes treatment.
Collapse
Affiliation(s)
- Patrick E. Gelbach
- Department of Biomedical Engineering, USC, Los Angeles, California, United States of America
| | - Dongqing Zheng
- Mork Family Department of Chemical Engineering and Materials Science, USC, Los Angeles, California, United States of America
| | - Scott E. Fraser
- Translational Imaging Center, University of Southern California, Los Angeles, California, United States of America
| | - Kate L. White
- Departments of Biological Sciences and Chemistry, Bridge Institute, USC Michelson Center, USC, Los Angeles, California, United States of America
| | - Nicholas A. Graham
- Mork Family Department of Chemical Engineering and Materials Science, USC, Los Angeles, California, United States of America
| | - Stacey D. Finley
- Department of Biomedical Engineering, USC, Los Angeles, California, United States of America
- Mork Family Department of Chemical Engineering and Materials Science, USC, Los Angeles, California, United States of America
- Department of Quantitative and Computational Biology, USC, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
8
|
Liu H, Zhang J, Wei C, Liu Z, Zhou W, Yang P, Gong Y, Zhao Y. Prognostic signature construction of energy metabolism-related genes in pancreatic cancer. Front Oncol 2022; 12:917897. [PMID: 36248974 PMCID: PMC9559226 DOI: 10.3389/fonc.2022.917897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic cancer is the 7th leading cause of cancer death worldwide, and its incidence and mortality rate have been on the rise in recent years in Western developed countries. The specificity of the disease and the lack of appropriate treatments have resulted in a 5-year overall survival rate of only 9%. In this study, we conducted a study based on the TCGA database and GEO database and analyzed using the energy metabolism gene set to establish a prognostic model with the least absolute shrinkage and selection operator to identify 7-genes prognostic signature, and the gene expression was verified by Real-time PCR. The model was validated using a risk score calculation, and the OS rates of the 7 genes were analyzed using one-way Cox regression. The prognostic relationship between vesicle-associated membrane protein 2 (VAMP2) and pancreatic cancer patients was analyzed by OS and progression-free survival, and the prognosis was found to be significantly worse in the high-expression group. A Nomogram showed that VAMP2 was an independent prognostic factor in pancreatic cancer. Gene set enrichment analysis showed that VAMP2 upregulation was enriched in pathways associated with immune response and that VAMP2 downregulation was enriched in metabolism-related pathways. The association of VAMP2 with immune cell infiltration was analyzed for the enrichment results, and VAMP2 was found to be positively associated with all 6 immune cells. The results of this study suggest that VAMP2 is an independent prognostic factor associated with energy metabolism in pancreatic cancer and may be involved in the immune response.
Collapse
Affiliation(s)
- Hao Liu
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
- United New Drug Research and Development Center, Hunan Biotrans Technology Co., LTD., Changsha, China
- Institute of Bioengineering, Biotrans Technology Co., LTD., Shanghai, China
| | - Jianhua Zhang
- Institute of Bioengineering, Biotrans Technology Co., LTD., Shanghai, China
| | - Chaoguang Wei
- Institute of Bioengineering, Biotrans Technology Co., LTD., Shanghai, China
| | - Zhao Liu
- United New Drug Research and Development Center, Hunan Biotrans Technology Co., LTD., Changsha, China
| | - Wei Zhou
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Pan Yang
- United New Drug Research and Development Center, Hunan Biotrans Technology Co., LTD., Changsha, China
| | - Yifu Gong
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
- *Correspondence: Yuxiang Zhao, ; Yifu Gong,
| | - Yuxiang Zhao
- United New Drug Research and Development Center, Hunan Biotrans Technology Co., LTD., Changsha, China
- Institute of Bioengineering, Biotrans Technology Co., LTD., Shanghai, China
- *Correspondence: Yuxiang Zhao, ; Yifu Gong,
| |
Collapse
|
9
|
Islam MM, Goertzen A, Singh PK, Saha R. Exploring the metabolic landscape of pancreatic ductal adenocarcinoma cells using genome-scale metabolic modeling. iScience 2022; 25:104483. [PMID: 35712079 PMCID: PMC9194136 DOI: 10.1016/j.isci.2022.104483] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/08/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major research focus because of its poor therapy response and dismal prognosis. PDAC cells adapt their metabolism to the surrounding environment, often relying on diverse nutrient sources. Because traditional experimental techniques appear exhaustive to find a viable therapeutic strategy, a highly curated and omics-informed PDAC genome-scale metabolic model was reconstructed using patient-specific transcriptomics data. From the model-predictions, several new metabolic functions were explored as potential therapeutic targets in addition to the known metabolic hallmarks of PDAC. Significant downregulation in the peroxisomal beta oxidation pathway, flux modulation in the carnitine shuttle system, and upregulation in the reactive oxygen species detoxification pathway reactions were observed. These unique metabolic traits of PDAC were correlated with potential drug combinations targeting genes with poor prognosis in PDAC. Overall, this study provides a better understanding of the metabolic vulnerabilities in PDAC and will lead to novel effective therapeutic strategies.
Collapse
Affiliation(s)
- Mohammad Mazharul Islam
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Andrea Goertzen
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Pankaj K. Singh
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Rajib Saha
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
10
|
Modeling of Tumor Growth with Input from Patient-Specific Metabolomic Data. Ann Biomed Eng 2022; 50:314-329. [PMID: 35083584 PMCID: PMC9743982 DOI: 10.1007/s10439-022-02904-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/01/2022] [Indexed: 12/15/2022]
Abstract
Advances in omic technologies have provided insight into cancer progression and treatment response. However, the nonlinear characteristics of cancer growth present a challenge to bridge from the molecular- to the tissue-scale, as tumor behavior cannot be encapsulated by the sum of the individual molecular details gleaned experimentally. Mathematical modeling and computational simulation have been traditionally employed to facilitate analysis of nonlinear systems. In this study, for the first time tumor metabolomic data are linked via mathematical modeling to the tumor tissue-scale behavior, showing the capability to mechanistically simulate cancer progression personalized to omic information obtainable from patient tumor core biopsy analysis. Generally, a higher degree of metabolic dysregulation has been correlated with more aggressive tumor behavior. Accordingly, key parameters influenced by metabolomic data in this model include tumor proliferation, vascularization, aggressiveness, lactic acid production, monocyte infiltration and macrophage polarization, and drug effect. The model enables evaluating interactions of interest between these parameters which drive tumor growth based on the metabolomic data. The results show that the model can group patients consistently with the clinically observed outcomes of response/non-response to chemotherapy. This modeling approach provides a first step towards evaluation of tumor growth based on tumor-specific metabolomic data.
Collapse
|
11
|
Luo M, Ma X, Jiang W, Zhang J, Liu W, Wei S, Liu H. Novel phosphanegold(I) thiolate complexes suppress de novo lipid synthesis in human lung cancer. Eur J Med Chem 2022; 232:114168. [DOI: 10.1016/j.ejmech.2022.114168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/17/2022] [Accepted: 01/30/2022] [Indexed: 12/13/2022]
|
12
|
Buchsbaum JC, Jaffray DA, Ba D, Borkon LL, Chalk C, Chung C, Coleman MA, Coleman CN, Diehn M, Droegemeier KK, Enderling H, Espey MG, Greenspan EJ, Hartshorn CM, Hoang T, Hsiao HT, Keppel C, Moore NW, Prior F, Stahlberg EA, Tourassi G, Willcox KE. Predictive Radiation Oncology - A New NCI-DOE Scientific Space and Community. Radiat Res 2022; 197:434-445. [PMID: 35090025 DOI: 10.1667/rade-22-00012.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/10/2022] [Indexed: 11/03/2022]
Abstract
With a widely attended virtual kickoff event on January 29, 2021, the National Cancer Institute (NCI) and the Department of Energy (DOE) launched a series of 4 interactive, interdisciplinary workshops-and a final concluding "World Café" on March 29, 2021-focused on advancing computational approaches for predictive oncology in the clinical and research domains of radiation oncology. These events reflect 3,870 human hours of virtual engagement with representation from 8 DOE national laboratories and the Frederick National Laboratory for Cancer Research (FNL), 4 research institutes, 5 cancer centers, 17 medical schools and teaching hospitals, 5 companies, 5 federal agencies, 3 research centers, and 27 universities. Here we summarize the workshops by first describing the background for the workshops. Participants identified twelve key questions-and collaborative parallel ideas-as the focus of work going forward to advance the field. These were then used to define short-term and longer-term "Blue Sky" goals. In addition, the group determined key success factors for predictive oncology in the context of radiation oncology, if not the future of all of medicine. These are: cross-discipline collaboration, targeted talent development, development of mechanistic mathematical and computational models and tools, and access to high-quality multiscale data that bridges mechanisms to phenotype. The workshop participants reported feeling energized and highly motivated to pursue next steps together to address the unmet needs in radiation oncology specifically and in cancer research generally and that NCI and DOE project goals align at the convergence of radiation therapy and advanced computing.
Collapse
Affiliation(s)
| | - David A Jaffray
- The University of Texas, MD Anderson Cancer Center, Houston, Texas 77030
| | - Demba Ba
- Harvard University, Cambridge, Massachusetts 02138
| | - Lynn L Borkon
- Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21701
| | | | - Caroline Chung
- The University of Texas, MD Anderson Cancer Center, Houston, Texas 77030
| | | | | | | | | | - Heiko Enderling
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | | | | | | | - Thuc Hoang
- U.S. Department of Energy, Washington, DC 20585
| | - H Timothy Hsiao
- American Society for Radiation Oncology (ASTRO), Arlington, Virginia 22202
| | | | - Nathan W Moore
- Sandia National Laboratories, Albuquerque, New Mexico 87123
| | - Fred Prior
- University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Eric A Stahlberg
- Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21701
| | | | | |
Collapse
|
13
|
Mertins SD. Capturing Biomarkers and Molecular Targets in Cellular Landscapes From Dynamic Reaction Network Models and Machine Learning. Front Oncol 2022; 11:805592. [PMID: 35127516 PMCID: PMC8813744 DOI: 10.3389/fonc.2021.805592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/31/2021] [Indexed: 12/02/2022] Open
Abstract
Computational dynamic ODE models of cell function describing biochemical reactions have been created for decades, but on a small scale. Still, they have been highly effective in describing and predicting behaviors. For example, oscillatory phospho-ERK levels were predicted and confirmed in MAPK signaling encompassing both positive and negative feedback loops. These models typically were limited and not adapted to large datasets so commonly found today. But importantly, ODE models describe reaction networks in well-mixed systems representing the cell and can be simulated with ordinary differential equations that are solved deterministically. Stochastic solutions, which can account for noisy reaction networks, in some cases, also improve predictions. Today, dynamic ODE models rarely encompass an entire cell even though it might be expected that an upload of the large genomic, transcriptomic, and proteomic datasets may allow whole cell models. It is proposed here to combine output from simulated dynamic ODE models, completed with omics data, to discover both biomarkers in cancer a priori and molecular targets in the Machine Learning setting.
Collapse
Affiliation(s)
- Susan D. Mertins
- Department of Science, Mount St. Mary’s University, Emmitsburg, MD, United States
- Biomedical Informatics and Data Science Directorate, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Limited Liability Company (LLC), Frederick, MD, United States
- BioSystems Strategies, Limited Liability Company (LLC), Frederick, MD, United States
- *Correspondence: Susan D. Mertins,
| |
Collapse
|
14
|
Brummer AB, Yang X, Ma E, Gutova M, Brown CE, Rockne RC. Dose-dependent thresholds of dexamethasone destabilize CAR T-cell treatment efficacy. PLoS Comput Biol 2022; 18:e1009504. [PMID: 35081104 PMCID: PMC8820647 DOI: 10.1371/journal.pcbi.1009504] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/07/2022] [Accepted: 01/12/2022] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is potentially an effective targeted immunotherapy for glioblastoma, yet there is presently little known about the efficacy of CAR T-cell treatment when combined with the widely used anti-inflammatory and immunosuppressant glucocorticoid, dexamethasone. Here we present a mathematical model-based analysis of three patient-derived glioblastoma cell lines treated in vitro with CAR T-cells and dexamethasone. Advanced in vitro experimental cell killing assay technologies allow for highly resolved temporal dynamics of tumor cells treated with CAR T-cells and dexamethasone, making this a valuable model system for studying the rich dynamics of nonlinear biological processes with translational applications. We model the system as a nonautonomous, two-species predator-prey interaction of tumor cells and CAR T-cells, with explicit time-dependence in the clearance rate of dexamethasone. Using time as a bifurcation parameter, we show that (1) dexamethasone destabilizes coexistence equilibria between CAR T-cells and tumor cells in a dose-dependent manner and (2) as dexamethasone is cleared from the system, a stable coexistence equilibrium returns in the form of a Hopf bifurcation. With the model fit to experimental data, we demonstrate that high concentrations of dexamethasone antagonizes CAR T-cell efficacy by exhausting, or reducing the activity of CAR T-cells, and by promoting tumor cell growth. Finally, we identify a critical threshold in the ratio of CAR T-cell death to CAR T-cell proliferation rates that predicts eventual treatment success or failure that may be used to guide the dose and timing of CAR T-cell therapy in the presence of dexamethasone in patients. Bioengineering and gene-editing technologies have paved the way for advance immunotherapies that can target patient-specific tumor cells. One of these therapies, chimeric antigen receptor (CAR) T-cell therapy has recently shown promise in treating glioblastoma, an aggressive brain cancer often with poor patient prognosis. Dexamethasone is a commonly prescribed anti-inflammatory medication due to the health complications of tumor associated swelling in the brain. However, the immunosuppressant effects of dexamethasone on the immunotherapeutic CAR T-cells are not well understood. To address this issue, we use mathematical modeling to study in vitro dynamics of dexamethasone and CAR T-cells in three patient-derived glioblastoma cell lines. We find that in each cell line studied there is a threshold of tolerable dexamethasone concentration. Below this threshold, CAR T-cells are successful at eliminating the cancer cells, while above this threshold, dexamethasone critically inhibits CAR T-cell efficacy. Our modeling suggests that in the presence of high dexamethasone reduced CAR T-cell efficacy, or increased exhaustion, can occur and result in CAR T-cell treatment failure.
Collapse
Affiliation(s)
- Alexander B. Brummer
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail: (ABB); (CEB); (RCR)
| | - Xin Yang
- Department of Hematology and Hematopoietic Cell Translation and Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Eric Ma
- Department of Hematology and Hematopoietic Cell Translation and Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
| | - Christine E. Brown
- Department of Hematology and Hematopoietic Cell Translation and Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail: (ABB); (CEB); (RCR)
| | - Russell C. Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, United States of America
- * E-mail: (ABB); (CEB); (RCR)
| |
Collapse
|
15
|
Wang J, Delfarah A, Gelbach PE, Fong E, Macklin P, Mumenthaler SM, Graham NA, Finley SD. Elucidating tumor-stromal metabolic crosstalk in colorectal cancer through integration of constraint-based models and LC-MS metabolomics. Metab Eng 2021; 69:175-187. [PMID: 34838998 PMCID: PMC8818109 DOI: 10.1016/j.ymben.2021.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/07/2021] [Accepted: 11/09/2021] [Indexed: 12/28/2022]
Abstract
Colorectal cancer (CRC) is a major cause of morbidity and mortality in the United States. Tumor-stromal metabolic crosstalk in the tumor microenvironment promotes CRC development and progression, but exactly how stromal cells, in particular cancer-associated fibroblasts (CAFs), affect the metabolism of tumor cells remains unknown. Here we take a data-driven approach to investigate the metabolic interactions between CRC cells and CAFs, integrating constraint-based modeling and metabolomic profiling. Using metabolomics data, we perform unsteady-state parsimonious flux balance analysis to infer flux distributions for central carbon metabolism in CRC cells treated with or without CAF-conditioned media. We find that CAFs reprogram CRC metabolism through stimulation of glycolysis, the oxidative arm of the pentose phosphate pathway (PPP), and glutaminolysis, as well as inhibition of the tricarboxylic acid cycle. To identify potential therapeutic targets, we simulate enzyme knockouts and find that CAF-treated CRC cells are especially sensitive to inhibitions of hexokinase and glucose-6-phosphate, the rate limiting steps of glycolysis and oxidative PPP. Our work gives mechanistic insights into the metabolic interactions between CRC cells and CAFs and provides a framework for testing hypotheses towards CRC-targeted therapies.
Collapse
Affiliation(s)
- Junmin Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Alireza Delfarah
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Patrick E Gelbach
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Emma Fong
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA, 90064, USA
| | - Paul Macklin
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, 46202, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, Los Angeles, CA, 90064, USA; Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA; Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
16
|
Sommariva S, Caviglia G, Ravera S, Frassoni F, Benvenuto F, Tortolina L, Castagnino N, Parodi S, Piana M. Computational quantification of global effects induced by mutations and drugs in signaling networks of colorectal cancer cells. Sci Rep 2021; 11:19602. [PMID: 34599254 PMCID: PMC8486743 DOI: 10.1038/s41598-021-99073-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most deadly and commonly diagnosed tumors worldwide. Several genes are involved in its development and progression. The most frequent mutations concern APC, KRAS, SMAD4, and TP53 genes, suggesting that CRC relies on the concomitant alteration of the related pathways. However, with classic molecular approaches, it is not easy to simultaneously analyze the interconnections between these pathways. To overcome this limitation, recently these pathways have been included in a huge chemical reaction network (CRN) describing how information sensed from the environment by growth factors is processed by healthy colorectal cells. Starting from this CRN, we propose a computational model which simulates the effects induced by single or multiple concurrent mutations on the global signaling network. The model has been tested in three scenarios. First, we have quantified the changes induced on the concentration of the proteins of the network by a mutation in APC, KRAS, SMAD4, or TP53. Second, we have computed the changes in the concentration of p53 induced by up to two concurrent mutations affecting proteins upstreams in the network. Third, we have considered a mutated cell affected by a gain of function of KRAS, and we have simulated the action of Dabrafenib, showing that the proposed model can be used to determine the most effective amount of drug to be delivered to the cell. In general, the proposed approach displays several advantages, in that it allows to quantify the alteration in the concentration of the proteins resulting from a single or multiple given mutations. Moreover, simulations of the global signaling network of CRC may be used to identify new therapeutic targets, or to disclose unexpected interactions between the involved pathways.
Collapse
Affiliation(s)
- Sara Sommariva
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy.
| | - Giacomo Caviglia
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| | - Silvia Ravera
- Dipartimento di Medicina Sperimentale, Università di Genova, Via De Toni 14, 16132, Genoa, Italy
| | - Francesco Frassoni
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| | - Federico Benvenuto
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| | - Lorenzo Tortolina
- Dipartimento di Medicina Interna, Università di Genova, via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Nicoletta Castagnino
- Dipartimento di Medicina Interna, Università di Genova, via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Silvio Parodi
- Dipartimento di Medicina Interna, Università di Genova, via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Michele Piana
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| |
Collapse
|
17
|
Raveh B, Sun L, White KL, Sanyal T, Tempkin J, Zheng D, Bharath K, Singla J, Wang C, Zhao J, Li A, Graham NA, Kesselman C, Stevens RC, Sali A. Bayesian metamodeling of complex biological systems across varying representations. Proc Natl Acad Sci U S A 2021; 118:e2104559118. [PMID: 34453000 PMCID: PMC8536362 DOI: 10.1073/pnas.2104559118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Comprehensive modeling of a whole cell requires an integration of vast amounts of information on various aspects of the cell and its parts. To divide and conquer this task, we introduce Bayesian metamodeling, a general approach to modeling complex systems by integrating a collection of heterogeneous input models. Each input model can in principle be based on any type of data and can describe a different aspect of the modeled system using any mathematical representation, scale, and level of granularity. These input models are 1) converted to a standardized statistical representation relying on probabilistic graphical models, 2) coupled by modeling their mutual relations with the physical world, and 3) finally harmonized with respect to each other. To illustrate Bayesian metamodeling, we provide a proof-of-principle metamodel of glucose-stimulated insulin secretion by human pancreatic β-cells. The input models include a coarse-grained spatiotemporal simulation of insulin vesicle trafficking, docking, and exocytosis; a molecular network model of glucose-stimulated insulin secretion signaling; a network model of insulin metabolism; a structural model of glucagon-like peptide-1 receptor activation; a linear model of a pancreatic cell population; and ordinary differential equations for systemic postprandial insulin response. Metamodeling benefits from decentralized computing, while often producing a more accurate, precise, and complete model that contextualizes input models as well as resolves conflicting information. We anticipate Bayesian metamodeling will facilitate collaborative science by providing a framework for sharing expertise, resources, data, and models, as exemplified by the Pancreatic β-Cell Consortium.
Collapse
Affiliation(s)
- Barak Raveh
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94158
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem 9190416, Israel
| | - Liping Sun
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Kate L White
- Department of Biological Sciences, Bridge Institute, University of Southern California, Los Angeles, CA 90089
| | - Tanmoy Sanyal
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94158
| | - Jeremy Tempkin
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94158
| | - Dongqing Zheng
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Kala Bharath
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94158
| | - Jitin Singla
- Department of Biological Sciences, Bridge Institute, University of Southern California, Los Angeles, CA 90089
- Epstein Department of Industrial and Systems Engineering, The Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
- Information Science Institute, The Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Chenxi Wang
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jihui Zhao
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Angdi Li
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Carl Kesselman
- Epstein Department of Industrial and Systems Engineering, The Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
- Information Science Institute, The Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Raymond C Stevens
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
- Department of Biological Sciences, Bridge Institute, University of Southern California, Los Angeles, CA 90089
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158;
- Quantitative Biosciences Institute, University of California, San Francisco, CA 94158
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158
| |
Collapse
|
18
|
Sahu A, Blätke MA, Szymański JJ, Töpfer N. Advances in flux balance analysis by integrating machine learning and mechanism-based models. Comput Struct Biotechnol J 2021; 19:4626-4640. [PMID: 34471504 PMCID: PMC8382995 DOI: 10.1016/j.csbj.2021.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
The availability of multi-omics data sets and genome-scale metabolic models for various organisms provide a platform for modeling and analyzing genotype-to-phenotype relationships. Flux balance analysis is the main tool for predicting flux distributions in genome-scale metabolic models and various data-integrative approaches enable modeling context-specific network behavior. Due to its linear nature, this optimization framework is readily scalable to multi-tissue or -organ and even multi-organism models. However, both data and model size can hamper a straightforward biological interpretation of the estimated fluxes. Moreover, flux balance analysis simulates metabolism at steady-state and thus, in its most basic form, does not consider kinetics or regulatory events. The integration of flux balance analysis with complementary data analysis and modeling techniques offers the potential to overcome these challenges. In particular machine learning approaches have emerged as the tool of choice for data reduction and selection of most important variables in big data sets. Kinetic models and formal languages can be used to simulate dynamic behavior. This review article provides an overview of integrative studies that combine flux balance analysis with machine learning approaches, kinetic models, such as physiology-based pharmacokinetic models, and formal graphical modeling languages, such as Petri nets. We discuss the mathematical aspects and biological applications of these integrated approaches and outline challenges and future perspectives.
Collapse
Affiliation(s)
- Ankur Sahu
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Corrensstraße 3, 06466 Gatersleben, Germany
| | - Mary-Ann Blätke
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Corrensstraße 3, 06466 Gatersleben, Germany
| | - Jędrzej Jakub Szymański
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Corrensstraße 3, 06466 Gatersleben, Germany
| | - Nadine Töpfer
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Corrensstraße 3, 06466 Gatersleben, Germany
| |
Collapse
|
19
|
Corral-Jara KF, Rosas da Silva G, Fierro NA, Soumelis V. Modeling the Th17 and Tregs Paradigm: Implications for Cancer Immunotherapy. Front Cell Dev Biol 2021; 9:675099. [PMID: 34026764 PMCID: PMC8137995 DOI: 10.3389/fcell.2021.675099] [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: 03/02/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
CD4 + T cell differentiation is governed by gene regulatory and metabolic networks, with both networks being highly interconnected and able to adapt to external stimuli. Th17 and Tregs differentiation networks play a critical role in cancer, and their balance is affected by the tumor microenvironment (TME). Factors from the TME mediate recruitment and expansion of Th17 cells, but these cells can act with pro or anti-tumor immunity. Tregs cells are also involved in tumor development and progression by inhibiting antitumor immunity and promoting immunoevasion. Due to the complexity of the underlying molecular pathways, the modeling of biological systems has emerged as a promising solution for better understanding both CD4 + T cell differentiation and cancer cell behavior. In this review, we present a context-dependent vision of CD4 + T cell transcriptomic and metabolic network adaptability. We then discuss CD4 + T cell knowledge-based models to extract the regulatory elements of Th17 and Tregs differentiation in multiple CD4 + T cell levels. We highlight the importance of complementing these models with data from omics technologies such as transcriptomics and metabolomics, in order to better delineate existing Th17 and Tregs bifurcation mechanisms. We were able to recompilate promising regulatory components and mechanisms of Th17 and Tregs differentiation under normal conditions, which we then connected with biological evidence in the context of the TME to better understand CD4 + T cell behavior in cancer. From the integration of mechanistic models with omics data, the transcriptomic and metabolomic reprograming of Th17 and Tregs cells can be predicted in new models with potential clinical applications, with special relevance to cancer immunotherapy.
Collapse
Affiliation(s)
- Karla F. Corral-Jara
- Computational Systems Biology Team, Institut de Biologie de l’Ecole Normale Supérieure, CNRS UMR 8197, INSERM U1024, Ecole Normale Supérieure, PSL Research University, Paris, France
| | | | - Nora A. Fierro
- Department of Immunology, Biomedical Research Institute, National Autonomous University of Mexico, Mexico City, Mexico
| | - Vassili Soumelis
- Université de Paris, INSERM U976, France and AP-HP, Hôpital Saint-Louis, Immunology-Histocompatibility Department, Paris, France
| |
Collapse
|
20
|
Sommariva S, Caviglia G, Piana M. Gain and loss of function mutations in biological chemical reaction networks: a mathematical model with application to colorectal cancer cells. J Math Biol 2021; 82:55. [PMID: 33945019 PMCID: PMC8096774 DOI: 10.1007/s00285-021-01607-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 03/20/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022]
Abstract
This paper studies a system of Ordinary Differential Equations modeling a chemical reaction network and derives from it a simulation tool mimicking Loss of Function and Gain of Function mutations found in cancer cells. More specifically, from a theoretical perspective, our approach focuses on the determination of moiety conservation laws for the system and their relation with the corresponding stoichiometric surfaces. Then we show that Loss of Function mutations can be implemented in the model via modification of the initial conditions in the system, while Gain of Function mutations can be implemented by eliminating specific reactions. Finally, the model is utilized to examine in detail the G1-S phase of a colorectal cancer cell.
Collapse
Affiliation(s)
- Sara Sommariva
- Dipartimento di Matematica, Universitá di Genova, Via Dodecaneso, 35 16146, Genoa, Italy
| | - Giacomo Caviglia
- Dipartimento di Matematica, Universitá di Genova, Via Dodecaneso, 35 16146, Genoa, Italy
| | - Michele Piana
- Dipartimento di Matematica, Universitá di Genova, Via Dodecaneso, 35 16146, Genoa, Italy. .,CNR - SPIN GENOVA, Via Dodecaneso, 35 16146, Genoa, Italy.
| |
Collapse
|
21
|
Hinow P, Pinter G, Yan W, Wang SE. Modeling the bidirectional glutamine/ammonium conversion between cancer cells and cancer-associated fibroblasts. PeerJ 2021; 9:e10648. [PMID: 33520452 PMCID: PMC7811294 DOI: 10.7717/peerj.10648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Like in an ecosystem, cancer and other cells residing in the tumor microenvironment engage in various modes of interactions to buffer the negative effects of environmental changes. One such change is the consumption of common nutrients (such as glutamine/Gln) and the consequent accumulation of toxic metabolic byproducts (such as ammonium/NH4). Ammonium is a waste product of cellular metabolism whose accumulation causes cell stress. In tumors, it is known that it can be recycled into nutrients by cancer associated fibroblasts (CAFs). Here we present monoculture and coculture growth of cancer cells and CAFs on different substrates: glutamine and ammonium. We propose a mathematical model to aid our understanding. We find that cancer cells are able to survive on ammonium and recycle it to glutamine for limited periods of time. CAFs are able to even grow on ammonium. In coculture, the presence of CAFs results in an improved survival of cancer cells compared to their monoculture when exposed to ammonium. Interestingly, the ratio between the two cell populations is maintained under various concentrations of NH4, suggesting the ability of the mixed cell system to survive temporary metabolic stress and sustain the size and cell composition as a stable entity.
Collapse
Affiliation(s)
- Peter Hinow
- Department of Mathematical Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Gabriella Pinter
- Department of Mathematical Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Wei Yan
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Shizhen Emily Wang
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
22
|
Sali A. From integrative structural biology to cell biology. J Biol Chem 2021; 296:100743. [PMID: 33957123 PMCID: PMC8203844 DOI: 10.1016/j.jbc.2021.100743] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/09/2021] [Accepted: 04/30/2021] [Indexed: 12/16/2022] Open
Abstract
Integrative modeling is an increasingly important tool in structural biology, providing structures by combining data from varied experimental methods and prior information. As a result, molecular architectures of large, heterogeneous, and dynamic systems, such as the ∼52-MDa Nuclear Pore Complex, can be mapped with useful accuracy, precision, and completeness. Key challenges in improving integrative modeling include expanding model representations, increasing the variety of input data and prior information, quantifying a match between input information and a model in a Bayesian fashion, inventing more efficient structural sampling, as well as developing better model validation, analysis, and visualization. In addition, two community-level challenges in integrative modeling are being addressed under the auspices of the Worldwide Protein Data Bank (wwPDB). First, the impact of integrative structures is maximized by PDB-Development, a prototype wwPDB repository for archiving, validating, visualizing, and disseminating integrative structures. Second, the scope of structural biology is expanded by linking the wwPDB resource for integrative structures with archives of data that have not been generally used for structure determination but are increasingly important for computing integrative structures, such as data from various types of mass spectrometry, spectroscopy, optical microscopy, proteomics, and genetics. To address the largest of modeling problems, a type of integrative modeling called metamodeling is being developed; metamodeling combines different types of input models as opposed to different types of data to compute an output model. Collectively, these developments will facilitate the structural biology mindset in cell biology and underpin spatiotemporal mapping of the entire cell.
Collapse
Affiliation(s)
- Andrej Sali
- Research Collaboratory for Structural Bioinformatics Protein Data Bank, the Department of Bioengineering and Therapeutic Sciences, the Quantitative Biosciences Institute (QBI), and the Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
23
|
Dasgupta A, Bakshi A, Chowdhury N, De RK. A control theoretic three timescale model for analyzing energy management in mammalian cancer cells. Comput Struct Biotechnol J 2020; 19:477-508. [PMID: 33510857 PMCID: PMC7809419 DOI: 10.1016/j.csbj.2020.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 11/26/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023] Open
Abstract
Interaction among different pathways, such as metabolic, signaling and gene regulatory networks, of cellular system is responsible to maintain homeostasis in a mammalian cell. Malfunctioning of this cooperation may lead to many complex diseases, such as cancer and type 2 diabetes. Timescale differences among these pathways make their integration a daunting task. Metabolic, signaling and gene regulatory networks have three different timescales, such as, ultrafast, fast and slow respectively. The article deals with this problem by developing a support vector regression (SVR) based three timescale model with the application of genetic algorithm based nonlinear controller. The proposed model can successfully capture the nonlinear transient dynamics and regulations of such integrated biochemical pathway under consideration. Besides, the model is quite capable of predicting the effects of certain drug targets for many types of complex diseases. Here, energy and cell proliferation management of mammalian cancer cells have been explored and analyzed with the help of the proposed novel approach. Previous investigations including in silico/in vivo/in vitro experiments have validated the results (the regulations of glucose transporter 1 (glut1), hexokinase (HK), and hypoxia-inducible factor-1 α (HIF-1 α ) among others, and the switching of pyruvate kinase (M2 isoform) between dimer and tetramer) generated by this model proving its effectiveness. Subsequently, the model predicts the effects of six selected drug targets, such as, the deactivation of transketolase and glucose-6-phosphate isomerase among others, in the case of mammalian malignant cells in terms of growth, proliferation, fermentation, and energy supply in the form of adenosine triphosphate (ATP).
Collapse
Affiliation(s)
- Abhijit Dasgupta
- Department of Data Science, School of Interdisciplinary Studies, University of Kalyani, Kalyani, Nadia 741235, West Bengal, India
| | - Abhisek Bakshi
- Department of Information Technology, Bengal Institute of Technology, Basanti Highway, Kolkata 700150, India
| | - Nirmalya Chowdhury
- Department of Computer Science & Engineering, Jadavpur University, Kolkata 700032, India
| | - Rajat K. De
- Machine Intelligence Unit, Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| |
Collapse
|
24
|
Aghakhani S, Zerrouk N, Niarakis A. Metabolic Reprogramming of Fibroblasts as Therapeutic Target in Rheumatoid Arthritis and Cancer: Deciphering Key Mechanisms Using Computational Systems Biology Approaches. Cancers (Basel) 2020; 13:E35. [PMID: 33374292 PMCID: PMC7795338 DOI: 10.3390/cancers13010035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/29/2022] Open
Abstract
Fibroblasts, the most abundant cells in the connective tissue, are key modulators of the extracellular matrix (ECM) composition. These spindle-shaped cells are capable of synthesizing various extracellular matrix proteins and collagen. They also provide the structural framework (stroma) for tissues and play a pivotal role in the wound healing process. While they are maintainers of the ECM turnover and regulate several physiological processes, they can also undergo transformations responding to certain stimuli and display aggressive phenotypes that contribute to disease pathophysiology. In this review, we focus on the metabolic pathways of glucose and highlight metabolic reprogramming as a critical event that contributes to the transition of fibroblasts from quiescent to activated and aggressive cells. We also cover the emerging evidence that allows us to draw parallels between fibroblasts in autoimmune disorders and more specifically in rheumatoid arthritis and cancer. We link the metabolic changes of fibroblasts to the toxic environment created by the disease condition and discuss how targeting of metabolic reprogramming could be employed in the treatment of such diseases. Lastly, we discuss Systems Biology approaches, and more specifically, computational modeling, as a means to elucidate pathogenetic mechanisms and accelerate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Sahar Aghakhani
- GenHotel, University of Evry, University of Paris-Saclay, Genopole, 91000 Evry, France; (S.A.); (N.Z.)
- Lifeware Group, Inria Saclay, 91120 Palaiseau, France
| | - Naouel Zerrouk
- GenHotel, University of Evry, University of Paris-Saclay, Genopole, 91000 Evry, France; (S.A.); (N.Z.)
| | - Anna Niarakis
- GenHotel, University of Evry, University of Paris-Saclay, Genopole, 91000 Evry, France; (S.A.); (N.Z.)
- Lifeware Group, Inria Saclay, 91120 Palaiseau, France
| |
Collapse
|
25
|
Role of Glutathione in Cancer: From Mechanisms to Therapies. Biomolecules 2020; 10:biom10101429. [PMID: 33050144 PMCID: PMC7600400 DOI: 10.3390/biom10101429] [Citation(s) in RCA: 382] [Impact Index Per Article: 76.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/30/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022] Open
Abstract
Glutathione (GSH) is the most abundant non-protein thiol present at millimolar concentrations in mammalian tissues. As an important intracellular antioxidant, it acts as a regulator of cellular redox state protecting cells from damage caused by lipid peroxides, reactive oxygen and nitrogen species, and xenobiotics. Recent studies have highlighted the importance of GSH in key signal transduction reactions as a controller of cell differentiation, proliferation, apoptosis, ferroptosis and immune function. Molecular changes in the GSH antioxidant system and disturbances in GSH homeostasis have been implicated in tumor initiation, progression, and treatment response. Hence, GSH has both protective and pathogenic roles. Although in healthy cells it is crucial for the removal and detoxification of carcinogens, elevated GSH levels in tumor cells are associated with tumor progression and increased resistance to chemotherapeutic drugs. Recently, several novel therapies have been developed to target the GSH antioxidant system in tumors as a means for increased response and decreased drug resistance. In this comprehensive review we explore mechanisms of GSH functionalities and different therapeutic approaches that either target GSH directly, indirectly or use GSH-based prodrugs. Consideration is also given to the computational methods used to describe GSH related processes for in silico testing of treatment effects.
Collapse
|
26
|
Frieboes HB, Raghavan S, Godin B. Modeling of Nanotherapy Response as a Function of the Tumor Microenvironment: Focus on Liver Metastasis. Front Bioeng Biotechnol 2020; 8:1011. [PMID: 32974325 PMCID: PMC7466654 DOI: 10.3389/fbioe.2020.01011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME) presents a challenging barrier for effective nanotherapy-mediated drug delivery to solid tumors. In particular for tumors less vascularized than the surrounding normal tissue, as in liver metastases, the structure of the organ itself conjures with cancer-specific behavior to impair drug transport and uptake by cancer cells. Cells and elements in the TME of hypovascularized tumors play a key role in the process of delivery and retention of anti-cancer therapeutics by nanocarriers. This brief review describes the drug transport challenges and how they are being addressed with advanced in vitro 3D tissue models as well as with in silico mathematical modeling. This modeling complements network-oriented techniques, which seek to interpret intra-cellular relevant pathways and signal transduction within cells and with their surrounding microenvironment. With a concerted effort integrating experimental observations with computational analyses spanning from the molecular- to the tissue-scale, the goal of effective nanotherapy customized to patient tumor-specific conditions may be finally realized.
Collapse
Affiliation(s)
- Hermann B. Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, United States
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
- Center for Predictive Medicine, University of Louisville, Louisville, KY, United States
| | - Shreya Raghavan
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, TX, United States
- Developmental Therapeutics Program, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
27
|
Wu Q, Finley SD. Mathematical Model Predicts Effective Strategies to Inhibit VEGF-eNOS Signaling. J Clin Med 2020; 9:jcm9051255. [PMID: 32357492 PMCID: PMC7287924 DOI: 10.3390/jcm9051255] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/12/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022] Open
Abstract
The endothelial nitric oxide synthase (eNOS) signaling pathway in endothelial cells has multiple physiological significances. It produces nitric oxide (NO), an important vasodilator, and enables a long-term proliferative response, contributing to angiogenesis. This signaling pathway is mediated by vascular endothelial growth factor (VEGF), a pro-angiogenic species that is often targeted to inhibit tumor angiogenesis. However, inhibiting VEGF-mediated eNOS signaling can lead to complications such as hypertension. Therefore, it is important to understand the dynamics of eNOS signaling in the context of angiogenesis inhibitors. Thrombospondin-1 (TSP1) is an important angiogenic inhibitor that, through interaction with its receptor CD47, has been shown to redundantly inhibit eNOS signaling. However, the exact mechanisms of TSP1's inhibitory effects on this pathway remain unclear. To address this knowledge gap, we established a molecular-detailed mechanistic model to describe VEGF-mediated eNOS signaling, and we used the model to identify the potential intracellular targets of TSP1. In addition, we applied the predictive model to investigate the effects of several approaches to selectively target eNOS signaling in cells experiencing high VEGF levels present in the tumor microenvironment. This work generates insights for pharmacologic targets and therapeutic strategies to inhibit tumor angiogenesis signaling while avoiding potential side effects in normal vasoregulation.
Collapse
Affiliation(s)
- Qianhui Wu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA;
| | - Stacey D. Finley
- Department of Biomedical Engineering, Mork Family Department of Chemical Engineering and Materials Science, and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: ; Tel.: +1-213-740-8788
| |
Collapse
|
28
|
Li W, Wang J. Uncovering the Underlying Mechanisms of Cancer Metabolism through the Landscapes and Probability Flux Quantifications. iScience 2020; 23:101002. [PMID: 32276228 PMCID: PMC7150521 DOI: 10.1016/j.isci.2020.101002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/03/2019] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer metabolism is critical for understanding the mechanism of tumorigenesis, yet the understanding is still challenging. We studied gene-metabolism regulatory interactions and quantified the global driving forces for cancer-metabolism dynamics as the underlying landscape and probability flux. We uncovered four steady-state attractors: a normal state attractor, a cancer OXPHOS state attractor, a cancer glycolysis state attractor, and an intermediate cancer state attractor. We identified the key regulatory interactions through global sensitivity analysis based on the landscape topography. Different landscape topographies of glycolysis switch between normal cells and cancer cells were identified. We uncovered that the normal state to cancer state transformation is associated with the peaks of the probability flux and the thermodynamic dissipation, giving dynamical and thermodynamic origin of cancer formation. We found that cancer metabolism oscillations consume more energy to support cancer malignancy. This study provides a quantitative understanding of cancer metabolism and suggests a metabolic therapeutic strategy.
Collapse
Affiliation(s)
- Wenbo Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Jin Wang
- Department of Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, NY 11794-3400, USA.
| |
Collapse
|
29
|
Dogra P, Ramírez JR, Peláez MJ, Wang Z, Cristini V, Parasher G, Rawat M. Mathematical Modeling to Address Challenges in Pancreatic Cancer. Curr Top Med Chem 2020; 20:367-376. [PMID: 31893993 PMCID: PMC7279939 DOI: 10.2174/1568026620666200101095641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/10/2019] [Accepted: 10/20/2019] [Indexed: 12/30/2022]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is regarded as one of the most lethal cancer types for its challenges associated with early diagnosis and resistance to standard chemotherapeutic agents, thereby leading to a poor five-year survival rate. The complexity of the disease calls for a multidisciplinary approach to better manage the disease and improve the status quo in PDAC diagnosis, prognosis, and treatment. To this end, the application of quantitative tools can help improve the understanding of disease mechanisms, develop biomarkers for early diagnosis, and design patient-specific treatment strategies to improve therapeutic outcomes. However, such approaches have only been minimally applied towards the investigation of PDAC, and we review the current status of mathematical modeling works in this field.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Javier Ruiz Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - María J. Peláez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Applied Physics Graduate Program, Rice University, Houston, TX 77005, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Gulshan Parasher
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
30
|
Rockne RC, Hawkins-Daarud A, Swanson KR, Sluka JP, Glazier JA, Macklin P, Hormuth DA, Jarrett AM, Lima EABF, Tinsley Oden J, Biros G, Yankeelov TE, Curtius K, Al Bakir I, Wodarz D, Komarova N, Aparicio L, Bordyuh M, Rabadan R, Finley SD, Enderling H, Caudell J, Moros EG, Anderson ARA, Gatenby RA, Kaznatcheev A, Jeavons P, Krishnan N, Pelesko J, Wadhwa RR, Yoon N, Nichol D, Marusyk A, Hinczewski M, Scott JG. The 2019 mathematical oncology roadmap. Phys Biol 2019; 16:041005. [PMID: 30991381 PMCID: PMC6655440 DOI: 10.1088/1478-3975/ab1a09] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Whether the nom de guerre is Mathematical Oncology, Computational or Systems Biology, Theoretical Biology, Evolutionary Oncology, Bioinformatics, or simply Basic Science, there is no denying that mathematics continues to play an increasingly prominent role in cancer research. Mathematical Oncology-defined here simply as the use of mathematics in cancer research-complements and overlaps with a number of other fields that rely on mathematics as a core methodology. As a result, Mathematical Oncology has a broad scope, ranging from theoretical studies to clinical trials designed with mathematical models. This Roadmap differentiates Mathematical Oncology from related fields and demonstrates specific areas of focus within this unique field of research. The dominant theme of this Roadmap is the personalization of medicine through mathematics, modelling, and simulation. This is achieved through the use of patient-specific clinical data to: develop individualized screening strategies to detect cancer earlier; make predictions of response to therapy; design adaptive, patient-specific treatment plans to overcome therapy resistance; and establish domain-specific standards to share model predictions and to make models and simulations reproducible. The cover art for this Roadmap was chosen as an apt metaphor for the beautiful, strange, and evolving relationship between mathematics and cancer.
Collapse
Affiliation(s)
- Russell C Rockne
- Department of Computational and Quantitative Medicine, Division of Mathematical Oncology, City of Hope National Medical Center, Duarte, CA 91010, United States of America. Author to whom any correspondence should be addressed
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Roy M, Finley SD. Metabolic reprogramming dynamics in tumor spheroids: Insights from a multicellular, multiscale model. PLoS Comput Biol 2019; 15:e1007053. [PMID: 31185009 PMCID: PMC6588258 DOI: 10.1371/journal.pcbi.1007053] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 06/21/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022] Open
Abstract
Mathematical modeling provides the predictive ability to understand the metabolic reprogramming and complex pathways that mediate cancer cells’ proliferation. We present a mathematical model using a multiscale, multicellular approach to simulate avascular tumor growth, applied to pancreatic cancer. The model spans three distinct spatial and temporal scales. At the extracellular level, reaction diffusion equations describe nutrient concentrations over a span of seconds. At the cellular level, a lattice-based energy driven stochastic approach describes cellular phenomena including adhesion, proliferation, viability and cell state transitions, occurring on the timescale of hours. At the sub-cellular level, we incorporate a detailed kinetic model of intracellular metabolite dynamics on the timescale of minutes, which enables the cells to uptake and excrete metabolites and use the metabolites to generate energy and building blocks for cell growth. This is a particularly novel aspect of the model. Certain defined criteria for the concentrations of intracellular metabolites lead to cancer cell growth, proliferation or death. Overall, we model the evolution of the tumor in both time and space. Starting with a cluster of tumor cells, the model produces an avascular tumor that quantitatively and qualitatively mimics experimental measurements of multicellular tumor spheroids. Through our model simulations, we can investigate the response of individual intracellular species under a metabolic perturbation and investigate how that response contributes to the response of the tumor as a whole. The predicted response of intracellular metabolites under various targeted strategies are difficult to resolve with experimental techniques. Thus, the model can give novel predictions as to the response of the tumor as a whole, identifies potential therapies to impede tumor growth, and predicts the effects of those therapeutic strategies. In particular, the model provides quantitative insight into the dynamic reprogramming of tumor cells at the intracellular level in response to specific metabolic perturbations. Overall, the model is a useful framework to study targeted metabolic strategies for inhibiting tumor growth. Cancer cells expertly alter their metabolism in order to sustain growth, a hallmark of cancer. Quantitative details about this metabolic reprogramming are difficult to obtain without the use of predictive mathematical models. Here, we present a robust computational model of avascular tumor growth. The novel aspect of this work lies in the incorporation of a detailed model of the dynamics of metabolism within each individual cell, which directly influence growth of the multicellular tumor as a whole. We apply the model to simulate how the tumor grows in space and time and to predict how the tumor responds to targeted inhibition of specific intracellular metabolic reactions. Our results show, first-hand, the dynamic metabolic reprogramming that occurs in cancer cells. Specifically, the model provides insight into how the cells alter their metabolism to compensate for the loss of a nutrient by exploiting alternative pathways for continued tumor growth. Our work provides a quantitative tool for identifying the impact of cellular and sub-cellular features on the evolution of a tumor. This framework is useful for developing potential cancer therapies, complementing experimental studies.
Collapse
Affiliation(s)
- Mahua Roy
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, United States of America
| | - Stacey D. Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, United States of America
- Mork Family Department of Chemical Engineering and Materials Science; Department of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
Diebner HH, Zerjatke T, Griehl M, Roeder I. Metabolism is the tie: The Bertalanffy-type cancer growth model as common denominator of various modelling approaches. Biosystems 2018; 167:1-23. [PMID: 29605248 DOI: 10.1016/j.biosystems.2018.03.004] [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] [Received: 02/22/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 10/17/2022]
Abstract
Cancer or tumour growth has been addressed from a variety of mathematical modelling perspectives in the past. Examples are single variable growth models, reaction diffusion models, compartment models, individual cell-based models, clonal competition models, to name only a few. In this paper, we show that the so called Bertalanffy-type growth model is a macroscopic model variant that can be conceived as an optimal condensed modelling approach that to a high degree preserves complexity with respect to the aforementioned more complex modelling variants. The derivation of the Bertalanffy-type model is crucially based on features of metabolism. Therefore, this model contains a shape parameter that can be interpreted as a resource utilisation efficiency. This shape parameter reflects features that are usually captured in much more complex models. To be specific, the shape parameter is related to morphological structures of tumours, which in turn depend on metabolic conditions. We, furthermore, show that a single variable variant of the Bertalanffy-type model can straightforwardly be extended to a multiclonal competition model. Since competition is crucially based on available shared or clone-specific resources, the metabolism-based approach is an obvious candidate to capture clonal competition. Depending on the specific context, metabolic reprogramming or other oncogene driven changes either lead to a suppression of cancer cells or to an improved competition resulting in outgrowth of tumours. The parametrisation of the Bertalanffy-type growth model allows to account for this observed variety of cancer characteristics. The shape parameter, conceived as a classifier for healthy and oncogenic phenotypes, supplies a link to survival and evolutionary stability concepts discussed in demographic studies, such as opportunistic versus equilibrium strategies.
Collapse
Affiliation(s)
- Hans H Diebner
- Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Institute for Medical Informatics and Biometry, Fetscherstrasse 74, D-01307 Dresden, Germany.
| | - Thomas Zerjatke
- Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Institute for Medical Informatics and Biometry, Fetscherstrasse 74, D-01307 Dresden, Germany
| | - Max Griehl
- Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Institute for Medical Informatics and Biometry, Fetscherstrasse 74, D-01307 Dresden, Germany
| | - Ingo Roeder
- Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Institute for Medical Informatics and Biometry, Fetscherstrasse 74, D-01307 Dresden, Germany
| |
Collapse
|