1
|
Glazar DJ, Sahebjam S, Yu HHM, Chen DT, Bhandari M, Enderling H. A sample size analysis of a mathematical model of longitudinal tumor volume and progression-free survival for Bayesian individual dynamic predictions in recurrent high-grade glioma. CPT Pharmacometrics Syst Pharmacol 2024. [PMID: 39737733 DOI: 10.1002/psp4.13290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/12/2024] [Accepted: 11/22/2024] [Indexed: 01/01/2025] Open
Abstract
Patients with recurrent high-grade glioma (rHGG) have a poor prognosis with median progression-free survival (PFS) of <7 months. Responses to treatment are heterogenous, suggesting a clinical need for prognostic models. Bayesian data analysis can exploit individual patient follow-up imaging studies to adaptively predict the risk of progression. We propose a novel sample size analysis for Bayesian individual dynamic predictions and demonstrate proof of principle. We coupled a nonlinear mixed effects tumor growth inhibition model with a survival model. Longitudinal tumor volumes and time-to-progression were simulated for 2000 in silico rHGG patients. Bayesian individual dynamic predictions of PFS curves were evaluated using area under the receiver operating characteristic curve (AUC) and Brier skill score (BSS). We investigated the effects of sample size on AUC and BSS margins of error. A power law relationship was observed between sample size and margins of error of AUC and BSS. Sample size was also found to be negatively correlated with margins of error and landmark time. We explored the use of this sample size analysis as a clinical look-up table for prospective clinical trial design and retrospective clinical data analysis. Here, we motivate the application of Bayesian individual dynamic predictions as a clinical end point for clinical trial design. Doing so could aid in the development of study protocols with patient-specific adaptations (escalate or de-escalate dose or frequency of drug administration, increase or decrease the frequency of follow-up, or change therapeutic modality) according to patient-specific prognosis. Future developments of this approach will focus on further model development and validation.
Collapse
Affiliation(s)
- Daniel J Glazar
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Solmaz Sahebjam
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Johns Hopkins Sidney Kimmel Cancer Center, Sibley Memorial Hospital, Washington, DC, USA
| | - Hsiang-Husan M Yu
- Department of Radiation Oncology, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | - Menal Bhandari
- Department of Radiation Oncology, Loyola University, Chicago, Illinois, USA
| | - Heiko Enderling
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2
|
Wang A, Yan R, Li H, Sun X, Zhou W, Smith SR. A joint-threshold Filippov model describing the effect of intermittent androgen-deprivation therapy in controlling prostate cancer. Math Biosci 2024; 377:109301. [PMID: 39307299 DOI: 10.1016/j.mbs.2024.109301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
Intermittent androgen-deprivation therapy (IADT) can be beneficial to delay the occurrence of treatment resistance and cancer relapse compared to the standard continuous therapy. To study the effect of IADT in controlling prostate cancer, we developed a Filippov prostate cancer model with a joint threshold function: therapy is implemented once the total population of androgen-dependent cells (AC-Ds) and androgen-independent cells (AC-Is) is greater than the threshold value ET, and it is suspended once the population is less than ET. As the parameters vary, our model undergoes a series of sliding bifurcations, including boundary node, focus, saddle, saddle-node and tangency bifurcations. We also obtained the coexistence of one, two or three real equilibria and the bistability of two equilibria. Our results demonstrate that the population of AC-Is can be contained at a predetermined level if the initial population of AC-Is is less than this level, and we choose a suitable threshold value.
Collapse
Affiliation(s)
- Aili Wang
- School of Science, Xi'an University of Technology, Xi'an 710054, PR China.
| | - Rong Yan
- School of Mathematics and Information Science, Baoji University of Arts and Sciences, Baoji 721013, PR China
| | - Haixia Li
- School of Mathematics and Information Science, Baoji University of Arts and Sciences, Baoji 721013, PR China
| | - Xiaodan Sun
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Weike Zhou
- School of Mathematics, Northwest University, Xi'an, Shaanxi 710127, PR China
| | - Stacey R Smith
- Department of Mathematics and Faculty of Medicine, The University of Ottawa, Ottawa, ON K1N 6N5, Canada.
| |
Collapse
|
3
|
Scibilia KR, Schlicke P, Schneller F, Kuttler C. Predicting resistance and pseudoprogression: are minimalistic immunoediting mathematical models capable of forecasting checkpoint inhibitor treatment outcomes in lung cancer? Math Biosci 2024; 376:109287. [PMID: 39218211 DOI: 10.1016/j.mbs.2024.109287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The increased application of immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 in lung cancer treatment generates clinical need to reliably predict individual patients' treatment outcomes. METHODS To bridge the prediction gap, we examine four different mathematical models in the form of ordinary differential equations, including a novel delayed response model. We rigorously evaluate their individual and combined predictive capabilities with regard to the patients' progressive disease (PD) status through equal weighting of model-derived outcome probabilities. RESULTS Fitting the complete treatment course, the novel delayed response model (R2=0.938) outperformed the simplest model (R2=0.865). The model combination was able to reliably predict patient PD outcome with an overall accuracy of 77% (sensitivity = 70%, specificity = 81%), solely through calibration with primary tumor longest diameter measurements. It autonomously identified a subset of 51% of patients where predictions with an overall accuracy of 81% (sensitivity = 81%, specificity = 81%) can be achieved. All models significantly outperformed a fully data-driven machine learning-based approach. IMPLICATIONS These modeling approaches provide a dynamic baseline framework to support clinicians in treatment decisions by identifying different treatment outcome trajectories with already clinically available measurement data. LIMITATIONS AND FUTURE DIRECTIONS Conjoint application of the presented approach with other predictive tools and biomarkers, as well as further disease information (e.g. metastatic stage), could further enhance treatment outcome prediction. We believe the simple model formulations allow widespread adoption of the developed models to other cancer types. Similar models can easily be formulated for other treatment modalities.
Collapse
Affiliation(s)
- Kevin Robert Scibilia
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 3, Garching, 85747, Germany
| | - Pirmin Schlicke
- Department of Mathematics, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 3, Garching, 85747, Germany.
| | - Folker Schneller
- Department of Internal Medicine III, Klinikum Rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich, 81675, Germany
| | - Christina Kuttler
- Department of Mathematics, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 3, Garching, 85747, Germany
| |
Collapse
|
4
|
Salvioli M, Vandelaer L, Baena E, Schneider K, Cavill R, Staňková K. The effect of tumor composition on the success of adaptive therapy: The case of metastatic Castrate-Resistant Prostate Cancer. PLoS One 2024; 19:e0308173. [PMID: 39325718 PMCID: PMC11426540 DOI: 10.1371/journal.pone.0308173] [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: 03/22/2023] [Accepted: 07/18/2024] [Indexed: 09/28/2024] Open
Abstract
Prostate-specific antigen (PSA) is the most commonly used serum marker for prostate cancer. It plays a role in cancer detection, treatment monitoring, and more recently, in guiding adaptive therapy protocols, where treatment is alternated based on PSA levels. However, the relationship between PSA levels and tumor volume remains poorly understood. Empirical evidence suggests that different cancer cell types produce varying amounts of PSA. Despite this, current mathematical cancer models often assume either that all cell types contribute equally to PSA levels or that only certain subpopulations produce PSA at fixed rates. In this study, we compare Zhang et al.'s classical adaptive therapy protocol with the standard of care, which involves continuous maximum tolerable dose treatment, under different assumptions regarding PSA production. Specifically, we explore the possibility that testosterone-dependent, testosterone-producing, and testosterone-independent cells contribute to PSA production to varying degrees. We use the time to competitive release as a proxy for the time to disease progression. Our findings indicate that adaptive therapy consistently results in a longer time to competitive release compared to the standard of care, regardless of the assumptions about PSA production. However, when testosterone-independent cells are the sole PSA producers, Zhang et al.'s adaptive therapy protocol becomes inapplicable, as PSA levels never fall to half of their initial value, preventing therapy discontinuation. Additionally, we observe that the number and duration of treatment cycles in adaptive therapy are highly sensitive to assumptions about how much each cell type contributes to PSA production. Overall, our results emphasize the need for a deeper understanding of patient-specific PSA dynamics, which could enhance the effectiveness of adaptive therapy in prostate cancer treatment.
Collapse
Affiliation(s)
- Monica Salvioli
- Evolutionary Game Theory Lab, Faculty of Technology, Policy and Management, Delft University of Technology, Delft, The Netherlands
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, The Netherlands
| | - Len Vandelaer
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, The Netherlands
| | - Esther Baena
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Katharina Schneider
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, The Netherlands
| | - Rachel Cavill
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, The Netherlands
| | - Kateřina Staňková
- Evolutionary Game Theory Lab, Faculty of Technology, Policy and Management, Delft University of Technology, Delft, The Netherlands
| |
Collapse
|
5
|
Mangrum DS, Finley SD. Modeling the heterogeneous apoptotic response of caspase-mediated signaling in tumor cells. J Theor Biol 2024; 590:111857. [PMID: 38797470 DOI: 10.1016/j.jtbi.2024.111857] [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: 10/31/2023] [Revised: 02/01/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Resisting apoptosis is a hallmark of cancer. For this reason, it may be possible to force cancer cells to die by targeting components along the apoptotic signaling pathway. However, apoptosis signaling is challenging to understand due to dynamic and complex behaviors of ligands, receptors, and intracellular signaling components in response to cancer therapy. In this work, we forecast the apoptotic response based on the combined impact of these features. We expanded a previously established mathematical model of caspase-mediated apoptosis to include extracellular activation and receptor dynamics. In addition, three potential threshold values of caspase-3 necessary for the activation of apoptosis were selected to forecast which cells become apoptotic over time. We first vary ligand and receptor levels with the number of intracellular signaling proteins remaining consistent. Then, we vary the intracellular protein molecules in each simulated tumor cell to forecast the response of a heterogeneous population. By leveraging the benefits of computational modeling, we investigate the combined effect of several factors on the onset of apoptosis. This work provides quantitative insights for how the apoptotic signaling response can be forecasted, and precisely triggered, amongst heterogeneous cells via extracellular activation.
Collapse
Affiliation(s)
- Diamond S Mangrum
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Stacey D Finley
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; Department of Quantitative and Computational Biology, 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.
| |
Collapse
|
6
|
Lorenzo G, Ahmed SR, Hormuth DA, Vaughn B, Kalpathy-Cramer J, Solorio L, Yankeelov TE, Gomez H. Patient-Specific, Mechanistic Models of Tumor Growth Incorporating Artificial Intelligence and Big Data. Annu Rev Biomed Eng 2024; 26:529-560. [PMID: 38594947 DOI: 10.1146/annurev-bioeng-081623-025834] [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] [Indexed: 04/11/2024]
Abstract
Despite the remarkable advances in cancer diagnosis, treatment, and management over the past decade, malignant tumors remain a major public health problem. Further progress in combating cancer may be enabled by personalizing the delivery of therapies according to the predicted response for each individual patient. The design of personalized therapies requires the integration of patient-specific information with an appropriate mathematical model of tumor response. A fundamental barrier to realizing this paradigm is the current lack of a rigorous yet practical mathematical theory of tumor initiation, development, invasion, and response to therapy. We begin this review with an overview of different approaches to modeling tumor growth and treatment, including mechanistic as well as data-driven models based on big data and artificial intelligence. We then present illustrative examples of mathematical models manifesting their utility and discuss the limitations of stand-alone mechanistic and data-driven models. We then discuss the potential of mechanistic models for not only predicting but also optimizing response to therapy on a patient-specific basis. We describe current efforts and future possibilities to integrate mechanistic and data-driven models. We conclude by proposing five fundamental challenges that must be addressed to fully realize personalized care for cancer patients driven by computational models.
Collapse
Affiliation(s)
- Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, University of Texas, Austin, Texas, USA
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| | - Syed Rakin Ahmed
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
- Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard Medical School, Harvard University, Cambridge, Massachusetts, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David A Hormuth
- Livestrong Cancer Institutes, University of Texas, Austin, Texas, USA
- Oden Institute for Computational Engineering and Sciences, University of Texas, Austin, Texas, USA
| | - Brenna Vaughn
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | | | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Thomas E Yankeelov
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Biomedical Engineering, Department of Oncology, and Department of Diagnostic Medicine, University of Texas, Austin, Texas, USA
- Livestrong Cancer Institutes, University of Texas, Austin, Texas, USA
- Oden Institute for Computational Engineering and Sciences, University of Texas, Austin, Texas, USA
| | - Hector Gomez
- School of Mechanical Engineering and Purdue Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| |
Collapse
|
7
|
Vibishan B, B V H, Dey S. A resource-based mechanistic framework for castration-resistant prostate cancer (CRPC). J Theor Biol 2024; 587:111806. [PMID: 38574968 DOI: 10.1016/j.jtbi.2024.111806] [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] [Revised: 02/04/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
Cancer therapy often leads to the selective elimination of drug-sensitive cells from the tumour. This can favour the growth of cells resistant to the therapeutic agent, ultimately causing a tumour relapse. Castration-resistant prostate cancer (CRPC) is a well-characterised instance of this phenomenon. In CRPC, after systemic androgen deprivation therapy (ADT), a subset of drug-resistant cancer cells autonomously produce testosterone, thus enabling tumour regrowth. A previous theoretical study has shown that such a tumour relapse can be delayed by inhibiting the growth of drug-resistant cells using biotic competition from drug-sensitive cells. In this context, the centrality of resource dynamics to intra-tumour competition in the CRPC system indicates clear scope for the construction of theoretical models that can explicitly incorporate the underlying mechanisms of tumour ecology. In the current study, we use a modified logistic framework to model cell-cell interactions in terms of the production and consumption of resources. Our results show that steady state composition of CRPC can be understood as a composite function of the availability and utilisation efficiency of two resources-oxygen and testosterone. In particular, we show that the effect of changing resource availability or use efficiency is conditioned by their general abundance regimes. Testosterone typically functions in trace amounts and thus affects steady state behaviour of the CRPC system differently from oxygen, which is usually available at higher levels. Our data thus indicate that explicit consideration of resource dynamics can produce novel and useful mechanistic understanding of CRPC. Furthermore, such a modelling approach also incorporates variables into the system's description that can be directly measured in a clinical context. This is therefore a promising avenue of research in cancer ecology that could lead to therapeutic approaches that are more clearly rooted in the biology of CRPC.
Collapse
Affiliation(s)
- B Vibishan
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Pune, Maharashtra, India.
| | - Harshavardhan B V
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Pune, Maharashtra, India; IISc Mathematics Initiative, Indian Institute of Science, Bangalore, Karnataka, India.
| | - Sutirth Dey
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Pune, Maharashtra, India.
| |
Collapse
|
8
|
Gallagher K, Strobl MA, Park DS, Spoendlin FC, Gatenby RA, Maini PK, Anderson AR. Mathematical Model-Driven Deep Learning Enables Personalized Adaptive Therapy. Cancer Res 2024; 84:1929-1941. [PMID: 38569183 PMCID: PMC11148552 DOI: 10.1158/0008-5472.can-23-2040] [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: 08/16/2023] [Revised: 01/05/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024]
Abstract
Standard-of-care treatment regimens have long been designed for maximal cell killing, yet these strategies often fail when applied to metastatic cancers due to the emergence of drug resistance. Adaptive treatment strategies have been developed as an alternative approach, dynamically adjusting treatment to suppress the growth of treatment-resistant populations and thereby delay, or even prevent, tumor progression. Promising clinical results in prostate cancer indicate the potential to optimize adaptive treatment protocols. Here, we applied deep reinforcement learning (DRL) to guide adaptive drug scheduling and demonstrated that these treatment schedules can outperform the current adaptive protocols in a mathematical model calibrated to prostate cancer dynamics, more than doubling the time to progression. The DRL strategies were robust to patient variability, including both tumor dynamics and clinical monitoring schedules. The DRL framework could produce interpretable, adaptive strategies based on a single tumor burden threshold, replicating and informing optimal treatment strategies. The DRL framework had no knowledge of the underlying mathematical tumor model, demonstrating the capability of DRL to help develop treatment strategies in novel or complex settings. Finally, a proposed five-step pathway, which combined mechanistic modeling with the DRL framework and integrated conventional tools to improve interpretability compared with traditional "black-box" DRL models, could allow translation of this approach to the clinic. Overall, the proposed framework generated personalized treatment schedules that consistently outperformed clinical standard-of-care protocols. SIGNIFICANCE Generation of interpretable and personalized adaptive treatment schedules using a deep reinforcement framework that interacts with a virtual patient model overcomes the limitations of standardized strategies caused by heterogeneous treatment responses.
Collapse
Affiliation(s)
- Kit Gallagher
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Oxford, United Kingdom
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | | | - Derek S. Park
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Fabian C. Spoendlin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Oxford, United Kingdom
| | - Robert A. Gatenby
- Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Philip K. Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, Oxford, United Kingdom
| | | |
Collapse
|
9
|
Bergman DR, Wang Y, Trujillo E, Fernald AA, Li L, Pearson AT, Sweis RF, Jackson TL. Dysregulated FGFR3 signaling alters the immune landscape in bladder cancer and presents therapeutic possibilities in an agent-based model. Front Immunol 2024; 15:1358019. [PMID: 38515743 PMCID: PMC10954792 DOI: 10.3389/fimmu.2024.1358019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Bladder cancer is an increasingly prevalent global disease that continues to cause morbidity and mortality despite recent advances in treatment. Immune checkpoint inhibitors (ICI) and fibroblast growth factor receptor (FGFR)-targeted therapeutics have had modest success in bladder cancer when used as monotherapy. Emerging data suggests that the combination of these two therapies could lead to improved clinical outcomes, but the optimal strategy for combining these agents remains uncertain. Mathematical models, specifically agent-based models (ABMs), have shown recent successes in uncovering the multiscale dynamics that shape the trajectory of cancer. They have enabled the optimization of treatment methods and the identification of novel therapeutic strategies. To assess the combined effects of anti-PD-1 and anti-FGFR3 small molecule inhibitors (SMI) on tumor growth and the immune response, we built an ABM that captures key facets of tumor heterogeneity and CD8+ T cell phenotypes, their spatial interactions, and their response to therapeutic pressures. Our model quantifies how tumor antigenicity and FGFR3 activating mutations impact disease trajectory and response to anti-PD-1 antibodies and anti-FGFR3 SMI. We find that even a small population of weakly antigenic tumor cells bearing an FGFR3 mutation can render the tumor resistant to combination therapy. However, highly antigenic tumors can overcome therapeutic resistance mediated by FGFR3 mutation. The optimal therapy depends on the strength of the FGFR3 signaling pathway. Under certain conditions, ICI alone is optimal; in others, ICI followed by anti-FGFR3 therapy is best. These results indicate the need to quantify FGFR3 signaling and the fitness advantage conferred on bladder cancer cells harboring this mutation. This ABM approach may enable rationally designed treatment plans to improve clinical outcomes.
Collapse
Affiliation(s)
- Daniel R. Bergman
- Department of Mathematics, University of Michigan, Ann Arbor, MI, United States
| | - Yixuan Wang
- Department of Mathematics, University of Michigan, Ann Arbor, MI, United States
| | - Erica Trujillo
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, United States
| | - Anthony A. Fernald
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, United States
| | - Lie Li
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, United States
| | - Alexander T. Pearson
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, United States
| | - Randy F. Sweis
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, United States
| | | |
Collapse
|
10
|
Lorenzo G, Heiselman JS, Liss MA, Miga MI, Gomez H, Yankeelov TE, Reali A, Hughes TJ. A Pilot Study on Patient-specific Computational Forecasting of Prostate Cancer Growth during Active Surveillance Using an Imaging-informed Biomechanistic Model. CANCER RESEARCH COMMUNICATIONS 2024; 4:617-633. [PMID: 38426815 PMCID: PMC10906139 DOI: 10.1158/2767-9764.crc-23-0449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/15/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Active surveillance (AS) is a suitable management option for newly diagnosed prostate cancer, which usually presents low to intermediate clinical risk. Patients enrolled in AS have their tumor monitored via longitudinal multiparametric MRI (mpMRI), PSA tests, and biopsies. Hence, treatment is prescribed when these tests identify progression to higher-risk prostate cancer. However, current AS protocols rely on detecting tumor progression through direct observation according to population-based monitoring strategies. This approach limits the design of patient-specific AS plans and may delay the detection of tumor progression. Here, we present a pilot study to address these issues by leveraging personalized computational predictions of prostate cancer growth. Our forecasts are obtained with a spatiotemporal biomechanistic model informed by patient-specific longitudinal mpMRI data (T2-weighted MRI and apparent diffusion coefficient maps from diffusion-weighted MRI). Our results show that our technology can represent and forecast the global tumor burden for individual patients, achieving concordance correlation coefficients from 0.93 to 0.99 across our cohort (n = 7). In addition, we identify a model-based biomarker of higher-risk prostate cancer: the mean proliferation activity of the tumor (P = 0.041). Using logistic regression, we construct a prostate cancer risk classifier based on this biomarker that achieves an area under the ROC curve of 0.83. We further show that coupling our tumor forecasts with this prostate cancer risk classifier enables the early identification of prostate cancer progression to higher-risk disease by more than 1 year. Thus, we posit that our predictive technology constitutes a promising clinical decision-making tool to design personalized AS plans for patients with prostate cancer. SIGNIFICANCE Personalization of a biomechanistic model of prostate cancer with mpMRI data enables the prediction of tumor progression, thereby showing promise to guide clinical decision-making during AS for each individual patient.
Collapse
Affiliation(s)
- Guillermo Lorenzo
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas
| | - Jon S. Heiselman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Michael A. Liss
- Department of Urology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Michael I. Miga
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Vanderbilt Institute for Surgery and Engineering, Vanderbilt University, Nashville, Tennessee
- Department of Neurological Surgery, Radiology, and Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hector Gomez
- School of Mechanical Engineering, Weldon School of Biomedical Engineering, and Purdue Institute for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Thomas E. Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas
- Livestrong Cancer Institutes and Departments of Biomedical Engineering, Diagnostic Medicine, and Oncology, The University of Texas at Austin, Austin, Texas
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alessandro Reali
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| | - Thomas J.R. Hughes
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
11
|
Enderling H. Fractal calculus in tumor growth simulations: The proof is in the pudding. Biosystems 2024; 237:105141. [PMID: 38355079 DOI: 10.1016/j.biosystems.2024.105141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Mathematical modeling in oncology has a long history. Recently, mathematical models and their predictions have made inroads into prospective clinical trials with encouraging results. The goal of many such modeling efforts is to make predictions, either to clinician's choice therapy or into "optimal" therapy - often for individual patients. The mathematical oncology community rightfully puts great hope into predictive modeling and mechanistic digital twins - but with this great opportunity comes great responsibility. Mathematical models need to be rigorously calibrated and validated, and their predictive performance ascertained, before conclusions about predictions into the unknown can be drawn. The recent article "Modeling tumor growth using fractal calculus: Insights into tumor dynamics" (Golmankhaneh et al., 2023), applied fractal calculus to tumor growth data. In this short commentary, I raise concerns about the study design and interpretation. In its current form, this study is poised to put cancer patients at risk if interpreted as concluded by the authors.
Collapse
Affiliation(s)
- Heiko Enderling
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Lu Y, Chu Q, Li Z, Wang M, Gatenby R, Zhang Q. Deep reinforcement learning identifies personalized intermittent androgen deprivation therapy for prostate cancer. Brief Bioinform 2024; 25:bbae071. [PMID: 38493345 PMCID: PMC11174533 DOI: 10.1093/bib/bbae071] [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: 10/09/2023] [Revised: 01/11/2024] [Accepted: 02/03/2024] [Indexed: 03/18/2024] Open
Abstract
The evolution of drug resistance leads to treatment failure and tumor progression. Intermittent androgen deprivation therapy (IADT) helps responsive cancer cells compete with resistant cancer cells in intratumoral competition. However, conventional IADT is population-based, ignoring the heterogeneity of patients and cancer. Additionally, existing IADT relies on pre-determined thresholds of prostate-specific antigen to pause and resume treatment, which is not optimized for individual patients. To address these challenges, we framed a data-driven method in two steps. First, we developed a time-varied, mixed-effect and generative Lotka-Volterra (tM-GLV) model to account for the heterogeneity of the evolution mechanism and the pharmacokinetics of two ADT drugs Cyproterone acetate and Leuprolide acetate for individual patients. Then, we proposed a reinforcement-learning-enabled individualized IADT framework, namely, I$^{2}$ADT, to learn the patient-specific tumor dynamics and derive the optimal drug administration policy. Experiments with clinical trial data demonstrated that the proposed I$^{2}$ADT can significantly prolong the time to progression of prostate cancer patients with reduced cumulative drug dosage. We further validated the efficacy of the proposed methods with a recent pilot clinical trial data. Moreover, the adaptability of I$^{2}$ADT makes it a promising tool for other cancers with the availability of clinical data, where treatment regimens might need to be individualized based on patient characteristics and disease dynamics. Our research elucidates the application of deep reinforcement learning to identify personalized adaptive cancer therapy.
Collapse
Affiliation(s)
- Yitao Lu
- School of Data Science, City University of Hong Kong, Hong Kong SAR, China
| | - Qian Chu
- Department of Thoracic Oncology, Tongji Hospital, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Mengdi Wang
- Department of Electrical and Computer Engineering and the Center for Statistics and Machine Learning, Princeton University, 08544, NJ, U.S.A
| | - Robert Gatenby
- Department of Integrated Mathematical Oncology and the Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, 33612, FL, USA
| | - Qingpeng Zhang
- Musketeers Foundation Institute of Data Science and the Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
13
|
Arabameri A, Arab S. Understanding the Interplay of CAR-NK Cells and Triple-Negative Breast Cancer: Insights from Computational Modeling. Bull Math Biol 2024; 86:20. [PMID: 38240892 DOI: 10.1007/s11538-023-01247-z] [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: 07/22/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024]
Abstract
Chimeric antigen receptor (CAR)-engineered natural killer (NK) cells have recently emerged as a promising and safe alternative to CAR-T cells for targeting solid tumors. In the case of triple-negative breast cancer (TNBC), traditional cancer treatments and common immunotherapies have shown limited effectiveness. However, CAR-NK cells have been successfully employed to target epidermal growth factor receptor (EGFR) on TNBC cells, thereby enhancing the efficacy of immunotherapy. The effectiveness of CAR-NK-based immunotherapy is influenced by various factors, including the vaccination dose, vaccination pattern, and tumor immunosuppressive factors in the microenvironment. To gain insights into the dynamics and effects of CAR-NK-based immunotherapy, we propose a computational model based on experimental data and immunological theories. This model integrates an individual-based model that describes the interplay between the tumor and the immune system, along with an ordinary differential equation model that captures the variation of inflammatory cytokines. Computational results obtained from the proposed model shed light on the conditions necessary for initiating an effective anti-tumor response. Furthermore, global sensitivity analysis highlights the issue of low persistence of CAR-NK cells in vivo, which poses a significant challenge for the successful clinical application of these cells. Leveraging the model, we identify the optimal vaccination time, vaccination dose, and time interval between injections for maximizing therapeutic outcomes.
Collapse
Affiliation(s)
- Abazar Arabameri
- Department of Electrical Engineering, University of Zanjan, Zanjan, Iran.
| | - Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
14
|
Yankeelov TE, Hormuth DA, Lima EA, Lorenzo G, Wu C, Okereke LC, Rauch GM, Venkatesan AM, Chung C. Designing clinical trials for patients who are not average. iScience 2024; 27:108589. [PMID: 38169893 PMCID: PMC10758956 DOI: 10.1016/j.isci.2023.108589] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Abstract
The heterogeneity inherent in cancer means that even a successful clinical trial merely results in a therapeutic regimen that achieves, on average, a positive result only in a subset of patients. The only way to optimize an intervention for an individual patient is to reframe their treatment as their own, personalized trial. Toward this goal, we formulate a computational framework for performing personalized trials that rely on four mathematical techniques. First, mathematical models that can be calibrated with patient-specific data to make accurate predictions of response. Second, digital twins built on these models capable of simulating the effects of interventions. Third, optimal control theory applied to the digital twins to optimize outcomes. Fourth, data assimilation to continually update and refine predictions in response to therapeutic interventions. In this perspective, we describe each of these techniques, quantify their "state of readiness", and identify use cases for personalized clinical trials.
Collapse
Affiliation(s)
- Thomas E. Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, 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
- Division of Diagnostic Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, 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
| | - Ernesto A.B.F. Lima
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- Texas Advanced Computer Center, The University of Texas at Austin, Austin, TX 78712, USA
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Civil Engineering and Architecture, University of Pavia, 27100 Pavia, Italy
| | - Chengyue Wu
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lois C. Okereke
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Gaiane M. Rauch
- Department of Abdominal Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Aradhana M. Venkatesan
- Department of Abdominal Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
15
|
Strobl MAR, Gallaher J, Robertson-Tessi M, West J, Anderson ARA. Treatment of evolving cancers will require dynamic decision support. Ann Oncol 2023; 34:867-884. [PMID: 37777307 PMCID: PMC10688269 DOI: 10.1016/j.annonc.2023.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 10/02/2023] Open
Abstract
Cancer research has traditionally focused on developing new agents, but an underexplored question is that of the dose and frequency of existing drugs. Based on the modus operandi established in the early days of chemotherapies, most drugs are administered according to predetermined schedules that seek to deliver the maximum tolerated dose and are only adjusted for toxicity. However, we believe that the complex, evolving nature of cancer requires a more dynamic and personalized approach. Chronicling the milestones of the field, we show that the impact of schedule choice crucially depends on processes driving treatment response and failure. As such, cancer heterogeneity and evolution dictate that a one-size-fits-all solution is unlikely-instead, each patient should be mapped to the strategy that best matches their current disease characteristics and treatment objectives (i.e. their 'tumorscape'). To achieve this level of personalization, we need mathematical modeling. In this perspective, we propose a five-step 'Adaptive Dosing Adjusted for Personalized Tumorscapes (ADAPT)' paradigm to integrate data and understanding across scales and derive dynamic and personalized schedules. We conclude with promising examples of model-guided schedule personalization and a call to action to address key outstanding challenges surrounding data collection, model development, and integration.
Collapse
Affiliation(s)
- M A R Strobl
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa; Translational Hematology and Oncology Research, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - J Gallaher
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - M Robertson-Tessi
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - J West
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - A R A Anderson
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa.
| |
Collapse
|
16
|
Bhatt AS, Schabath MB, Hoogland AI, Jim HS, Brady-Nicholls R. Patient-Reported Outcomes as Interradiographic Predictors of Response in Non-Small Cell Lung Cancer. Clin Cancer Res 2023; 29:3142-3150. [PMID: 37233986 PMCID: PMC10425729 DOI: 10.1158/1078-0432.ccr-23-0396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/19/2023] [Accepted: 05/24/2023] [Indexed: 05/27/2023]
Abstract
PURPOSE Minimally invasive biomarkers have been used as important indicators of treatment response and progression in cancers such as prostate and ovarian. Unfortunately, all biomarkers are not prognostic in all cancer types and are often not routinely collected. Patient-reported outcomes (PRO) provide a non-obtrusive, personalized measure of a patient's quality of life and symptomatology, reported directly from the patient, and are increasingly collected as part of routine care. Previous literature has shown correlations between specific PROs (i.e., insomnia, fatigue) and overall survival. Although promising, these studies often only consider single time points and ignore patient-specific dynamic changes in individual PROs, which might be early predictors of treatment response or progression. EXPERIMENTAL DESIGN In this study, PRO dynamics were analyzed to determine if they could be used as interradiographic predictors of tumor volume changes among 85 patients with non-small cell lung cancer undergoing immunotherapy. PRO questionnaires and tumor volume scans were completed biweekly and monthly, respectively. Correlation and predictive analysis were conducted to identify specific PROs that could accurately predict patient response. RESULTS Changes in tumor volume over time were significantly correlated with dizziness (P < 0.005), insomnia (P < 0.05), and fatigue (P < 0.05). In addition, cumulative changes in insomnia could predict progressive disease with a 77% accuracy, on average 45 days prior to the next imaging scan. CONCLUSIONS This study presents the first time that patient-specific PRO dynamics have been considered to predict how individual patients will respond to treatment. This is an important first step in adapting treatment to improve response rates.
Collapse
Affiliation(s)
- Ambika S. Bhatt
- Department of Biostatistics, Gillings School of Global Public Health at the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Matthew B. Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aasha I. Hoogland
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Heather S.L. Jim
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Renee Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
17
|
Davarci OO, Yang EY, Viguerie A, Yankeelov TE, Lorenzo G. Dynamic parameterization of a modified SEIRD model to analyze and forecast the dynamics of COVID-19 outbreaks in the United States. ENGINEERING WITH COMPUTERS 2023:1-25. [PMID: 37362241 PMCID: PMC10129322 DOI: 10.1007/s00366-023-01816-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 03/24/2023] [Indexed: 06/28/2023]
Abstract
The rapid spread of the numerous outbreaks of the coronavirus disease 2019 (COVID-19) pandemic has fueled interest in mathematical models designed to understand and predict infectious disease spread, with the ultimate goal of contributing to the decision making of public health authorities. Here, we propose a computational pipeline that dynamically parameterizes a modified SEIRD (susceptible-exposed-infected-recovered-deceased) model using standard daily series of COVID-19 cases and deaths, along with isolated estimates of population-level seroprevalence. We test our pipeline in five heavily impacted states of the US (New York, California, Florida, Illinois, and Texas) between March and August 2020, considering two scenarios with different calibration time horizons to assess the update in model performance as new epidemiologic data become available. Our results show a median normalized root mean squared error (NRMSE) of 2.38% and 4.28% in calibrating cumulative cases and deaths in the first scenario, and 2.41% and 2.30% when new data are assimilated in the second scenario, respectively. Then, 2-week (4-week) forecasts of the calibrated model resulted in median NRMSE of cumulative cases and deaths of 5.85% and 4.68% (8.60% and 17.94%) in the first scenario, and 1.86% and 1.93% (2.21% and 1.45%) in the second. Additionally, we show that our method provides significantly more accurate predictions of cases and deaths than a constant parameterization in the second scenario (p < 0.05). Thus, we posit that our methodology is a promising approach to analyze the dynamics of infectious disease outbreaks, and that our forecasts could contribute to designing effective pandemic-arresting public health policies. Supplementary Information The online version contains supplementary material available at 10.1007/s00366-023-01816-9.
Collapse
Affiliation(s)
- Orhun O. Davarci
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E 24th St, Austin, TX 78712-1229 USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
| | - Emily Y. Yang
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E 24th St, Austin, TX 78712-1229 USA
| | | | - Thomas E. Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E 24th St, Austin, TX 78712-1229 USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX USA
- Department of Oncology, The University of Texas at Austin, Austin, TX USA
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, TX USA
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E 24th St, Austin, TX 78712-1229 USA
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
| |
Collapse
|
18
|
Plaszczynski S, Grammaticos B, Pallud J, Campagne JE, Badoual M. Predicting regrowth of low-grade gliomas after radiotherapy. PLoS Comput Biol 2023; 19:e1011002. [PMID: 37000852 PMCID: PMC10128962 DOI: 10.1371/journal.pcbi.1011002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 04/25/2023] [Accepted: 03/04/2023] [Indexed: 04/03/2023] Open
Abstract
Diffuse low grade gliomas are invasive and incurable brain tumors that inevitably transform into higher grade ones. A classical treatment to delay this transition is radiotherapy (RT). Following RT, the tumor gradually shrinks during a period of typically 6 months to 4 years before regrowing. To improve the patient’s health-related quality of life and help clinicians build personalized follow-ups, one would benefit from predictions of the time during which the tumor is expected to decrease. The challenge is to provide a reliable estimate of this regrowth time shortly after RT (i.e. with few data), although patients react differently to the treatment. To this end, we analyze the tumor size dynamics from a batch of 20 high-quality longitudinal data, and propose a simple and robust analytical model, with just 4 parameters. From the study of their correlations, we build a statistical constraint that helps determine the regrowth time even for patients for which we have only a few measurements of the tumor size. We validate the procedure on the data and predict the regrowth time at the moment of the first MRI after RT, with precision of, typically, 6 months. Using virtual patients, we study whether some forecast is still possible just three months after RT. We obtain some reliable estimates of the regrowth time in 75% of the cases, in particular for all “fast-responders”. The remaining 25% represent cases where the actual regrowth time is large and can be safely estimated with another measurement a year later. These results show the feasibility of making personalized predictions of the tumor regrowth time shortly after RT.
Collapse
Affiliation(s)
- Stéphane Plaszczynski
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, Orsay, France
- Université Paris-Cité, IJCLab, Orsay, France
- * E-mail:
| | - Basile Grammaticos
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, Orsay, France
- Université Paris-Cité, IJCLab, Orsay, France
| | - Johan Pallud
- Department of Neurosurgery, GHU Paris Sainte-Anne Hospital, Paris, France
- Université de Paris, Sorbonne Paris Cité, Paris, France
- Inserm, U1266, IMA-Brain, Institut de Psychiatrie et Neurosciences de Paris, Paris, France
| | - Jean-Eric Campagne
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, Orsay, France
- Université Paris-Cité, IJCLab, Orsay, France
| | - Mathilde Badoual
- Université Paris-Saclay, CNRS/IN2P3, IJCLab, Orsay, France
- Université Paris-Cité, IJCLab, Orsay, France
| |
Collapse
|
19
|
West J, Adler F, Gallaher J, Strobl M, Brady-Nicholls R, Brown J, Roberson-Tessi M, Kim E, Noble R, Viossat Y, Basanta D, Anderson ARA. A survey of open questions in adaptive therapy: Bridging mathematics and clinical translation. eLife 2023; 12:e84263. [PMID: 36952376 PMCID: PMC10036119 DOI: 10.7554/elife.84263] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023] Open
Abstract
Adaptive therapy is a dynamic cancer treatment protocol that updates (or 'adapts') treatment decisions in anticipation of evolving tumor dynamics. This broad term encompasses many possible dynamic treatment protocols of patient-specific dose modulation or dose timing. Adaptive therapy maintains high levels of tumor burden to benefit from the competitive suppression of treatment-sensitive subpopulations on treatment-resistant subpopulations. This evolution-based approach to cancer treatment has been integrated into several ongoing or planned clinical trials, including treatment of metastatic castrate resistant prostate cancer, ovarian cancer, and BRAF-mutant melanoma. In the previous few decades, experimental and clinical investigation of adaptive therapy has progressed synergistically with mathematical and computational modeling. In this work, we discuss 11 open questions in cancer adaptive therapy mathematical modeling. The questions are split into three sections: (1) integrating the appropriate components into mathematical models (2) design and validation of dosing protocols, and (3) challenges and opportunities in clinical translation.
Collapse
Affiliation(s)
- Jeffrey West
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Fred Adler
- Department of Mathematics, University of UtahSalt Lake CityUnited States
- School of Biological Sciences, University of UtahSalt Lake CityUnited States
| | - Jill Gallaher
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Maximilian Strobl
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Renee Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Joel Brown
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Mark Roberson-Tessi
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Eunjung Kim
- Natural Product Informatics Research Center, Korea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Robert Noble
- Department of Mathematics, University of LondonLondonUnited Kingdom
| | - Yannick Viossat
- Ceremade, Université Paris-Dauphine, Université Paris Sciences et LettresParisFrance
| | - David Basanta
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Alexander RA Anderson
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| |
Collapse
|
20
|
Phan T, Bennett J, Patten T. Practical Understanding of Cancer Model Identifiability in Clinical Applications. Life (Basel) 2023; 13:410. [PMID: 36836767 PMCID: PMC9961656 DOI: 10.3390/life13020410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/28/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Mathematical models are a core component in the foundation of cancer theory and have been developed as clinical tools in precision medicine. Modeling studies for clinical applications often assume an individual's characteristics can be represented as parameters in a model and are used to explain, predict, and optimize treatment outcomes. However, this approach relies on the identifiability of the underlying mathematical models. In this study, we build on the framework of an observing-system simulation experiment to study the identifiability of several models of cancer growth, focusing on the prognostic parameters of each model. Our results demonstrate that the frequency of data collection, the types of data, such as cancer proxy, and the accuracy of measurements all play crucial roles in determining the identifiability of the model. We also found that highly accurate data can allow for reasonably accurate estimates of some parameters, which may be the key to achieving model identifiability in practice. As more complex models required more data for identification, our results support the idea of using models with a clear mechanism that tracks disease progression in clinical settings. For such a model, the subset of model parameters associated with disease progression naturally minimizes the required data for model identifiability.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87544, USA
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Justin Bennett
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85281, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Taylor Patten
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85281, USA
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| |
Collapse
|
21
|
van Winden LJ, Lanfermeijer M, Dezentje V, Bergman AM, van der Poel HG, van Rossum HH. Serum testosterone measured by liquid chromatography-tandem mass spectrometry is an independent predictor of response to castration in metastatic hormone-sensitive prostate cancer. Clin Chim Acta 2023; 539:34-40. [PMID: 36460134 DOI: 10.1016/j.cca.2022.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Although testosterone levels have been associated with progression-free survival (PFS) in metastatic hormone-sensitive prostate cancer (mHSPC) patients, this has primarily been investigated using inaccurate immunoassays (IA). Here, we investigated whether castrate testosterone levels determined by a liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay is an independent risk factor for treatment response in mHSPC. METHODS In total, 106 mHSPC patients treated with luteinizing-hormone releasing-hormone (LHRH) agonists were retrospectively analyzed between March 2018 and August 2021. Testosterone levels in serum samples were quantitated using an LC-MS/MS assay. In a subset of patients, IA (Roche Cobas Pro) values were compared with LC-MS/MS results. Survival analysis was performed using Kaplan-Meier curves and Cox proportional hazard models. RESULTS Median PFS was shorter for high testosterone levels (>0.231 nmol/L, 18.4 v. 42.6 months, HR 1.7, p = 0.018). Low testosterone levels and a PSA response below 4 ng/mL was associated with longer median PFS (46.2 months) than the remaining combinations (13.8-19.3 months, 3.4-5.8, overall p < 0.01). In 67 patients, testosterone levels below the median remained associated with longer PFS, whereas IA measurements did not show a similar difference. CONCLUSION Our results suggest that high castration testosterone levels measured by LC-MS/MS is an independent response predictor for mHSPC patients.
Collapse
Affiliation(s)
- Lennart J van Winden
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Mirthe Lanfermeijer
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vincent Dezentje
- Departments of Medical Oncology and Oncogenomics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andries M Bergman
- Departments of Medical Oncology and Oncogenomics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Urology, Amsterdam medical centers, Amsterdam, the Netherlands
| | - Huub H van Rossum
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Lorenzo G, di Muzio N, Deantoni CL, Cozzarini C, Fodor A, Briganti A, Montorsi F, Pérez-García VM, Gomez H, Reali A. Patient-specific forecasting of postradiotherapy prostate-specific antigen kinetics enables early prediction of biochemical relapse. iScience 2022; 25:105430. [DOI: 10.1016/j.isci.2022.105430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 09/04/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
|
23
|
Brady-Nicholls R, Enderling H. Range-Bounded Adaptive Therapy in Metastatic Prostate Cancer. Cancers (Basel) 2022; 14:5319. [PMID: 36358738 PMCID: PMC9657943 DOI: 10.3390/cancers14215319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 03/31/2024] Open
Abstract
Adaptive therapy with abiraterone acetate (AA), whereby treatment is cycled on and off, has been presented as an alternative to continuous therapy for metastatic castration resistant prostate cancer (mCRPC). It is hypothesized that cycling through treatment allows sensitive cells to competitively suppress resistant cells, thereby increasing the amount of time that treatment is effective. It has been proposed that there exists a subset of patients for whom this competition can be enhanced through slight modifications. Here, we investigate how adaptive AA can be modified to extend time to progression using a simple mathematical model of stem cell, non-stem cell, and prostate-specific antigen (PSA) dynamics. The model is calibrated to longitudinal PSA data from 16 mCRPC patients undergoing adaptive AA in a pilot clinical study at Moffitt Cancer Center. Model parameters are then used to simulate range-bounded adaptive therapy (RBAT) whereby treatment is modulated to maintain PSA levels between pre-determined patient-specific bounds. Model simulations of RBAT are compared to the clinically applied adaptive therapy and show that RBAT can further extend time to progression, while reducing the cumulative dose patients received in 11/16 patients. Simulations also show that the cumulative dose can be reduced by up to 40% under RBAT. Through small modifications to the conventional adaptive therapy design, our study demonstrates that RBAT offers the opportunity to improve patient care, particularly in those patients who do not respond well to conventional adaptive therapy.
Collapse
Affiliation(s)
- Renee Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
24
|
Shyntar A, Patel A, Rhodes M, Enderling H, Hillen T. The Tumor Invasion Paradox in Cancer Stem Cell-Driven Solid Tumors. Bull Math Biol 2022; 84:139. [PMID: 36301402 PMCID: PMC9613767 DOI: 10.1007/s11538-022-01086-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022]
Abstract
Cancer stem cells (CSCs) are key in understanding tumor growth and tumor progression. A counterintuitive effect of CSCs is the so-called tumor growth paradox: the effect where a tumor with a higher death rate may grow larger than a tumor with a lower death rate. Here we extend the modeling of the tumor growth paradox by including spatial structure and considering cancer invasion. Using agent-based modeling and a corresponding partial differential equation model, we demonstrate and prove mathematically a tumor invasion paradox: a larger cell death rate can lead to a faster invasion speed. We test this result on a generic hypothetical cancer with typical growth rates and typical treatment sensitivities. We find that the tumor invasion paradox may play a role for continuous and intermittent treatments, while it does not seem to be essential in fractionated treatments. It should be noted that no attempt was made to fit the model to a specific cancer, thus, our results are generic and theoretical.
Collapse
|
25
|
Zhang J, Gallaher J, Cunningham JJ, Choi JW, Ionescu F, Chatwal MS, Jain R, Kim Y, Wang L, Brown JS, Anderson AR, Gatenby RA. A Phase 1b Adaptive Androgen Deprivation Therapy Trial in Metastatic Castration Sensitive Prostate Cancer. Cancers (Basel) 2022; 14:5225. [PMID: 36358643 PMCID: PMC9656891 DOI: 10.3390/cancers14215225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background: We hypothesize that cancer survival can be improved through adapting treatment strategies to cancer evolutionary dynamics and conducted a phase 1b study in metastatic castration sensitive prostate cancer (mCSPC). Methods: Men with asymptomatic mCSPC were enrolled and proceeded with a treatment break after achieving > 75% PSA decline with LHRH analog plus an NHA. ADT was restarted at the time of PSA or radiographic progression and held again after achieving >50% PSA decline. This on-off cycling of ADT continued until on treatment imaging progression. Results: At data cut off in August 2022, only 2 of the 16 evaluable patients were off study due to imaging progression at 28 months from first dose of LHRH analog for mCSPC. Two additional patients showed PSA progression at 12.4 and 20.5 months and remain on trial. Since none of the 16 patients developed imaging progression at 12 months, the study succeeded in its primary objective of feasibility. The secondary endpoints of median time to PSA progression and median time to radiographic progression have not been reached at a median follow up of 26 months. Conclusions: It is feasible to use an individual’s PSA response and testosterone levels to guide intermittent ADT in mCSPC.
Collapse
Affiliation(s)
- Jingsong Zhang
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jill Gallaher
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | - Jung W. Choi
- Department of Radiology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Filip Ionescu
- Department of Oncological Science, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Monica S. Chatwal
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Rohit Jain
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Liang Wang
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Joel S. Brown
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Alexander R. Anderson
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Robert A. Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Department of Radiology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
26
|
Meade W, Weber A, Phan T, Hampston E, Resa LF, Nagy J, Kuang Y. High Accuracy Indicators of Androgen Suppression Therapy Failure for Prostate Cancer-A Modeling Study. Cancers (Basel) 2022; 14:cancers14164033. [PMID: 36011026 PMCID: PMC9406554 DOI: 10.3390/cancers14164033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Hormonal therapy for prostate cancer is often applied past the point of resistance, hence losing any future clinical value to the evolution of resistant strains. If the undesirable outcome of the treatment is forewarned, then clinicians can have an opportunity to adjust the treatment, which can result in better management of the cancer. Using a mechanistic mathematical model, we introduce two methods to enhance the accuracy of classical biomarkers for hormonal therapy failure. Our results show the value in measuring both prostate-specific antigen and androgen during hormonal treatment, which can potentially allow for better management of prostate cancer. Abstract Prostate cancer is a serious public health concern in the United States. The primary obstacle to effective long-term management for prostate cancer patients is the eventual development of treatment resistance. Due to the uniquely chaotic nature of the neoplastic genome, it is difficult to determine the evolution of tumor composition over the course of treatment. Hence, a drug is often applied continuously past the point of effectiveness, thereby losing any potential treatment combination with that drug permanently to resistance. If a clinician is aware of the timing of resistance to a particular drug, then they may have a crucial opportunity to adjust the treatment to retain the drug’s usefulness in a potential treatment combination or strategy. In this study, we investigate new methods of predicting treatment failure due to treatment resistance using a novel mechanistic model built on an evolutionary interpretation of Droop cell quota theory. We analyze our proposed methods using patient PSA and androgen data from a clinical trial of intermittent treatment with androgen deprivation therapy. Our results produce two indicators of treatment failure. The first indicator, proposed from the evolutionary nature of the cancer population, is calculated using our mathematical model with a predictive accuracy of 87.3% (sensitivity: 96.1%, specificity: 65%). The second indicator, conjectured from the implication of the first indicator, is calculated directly from serum androgen and PSA data with a predictive accuracy of 88.7% (sensitivity: 90.2%, specificity: 85%). Our results demonstrate the potential and feasibility of using an evolutionary tumor dynamics model in combination with the appropriate data to aid in the adaptive management of prostate cancer.
Collapse
Affiliation(s)
- William Meade
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Allison Weber
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ 85281, USA
- College of Computing, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Tin Phan
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Emily Hampston
- Department of Mathematics, State University of New York, Buffalo, NY 14260, USA
| | - Laura Figueroa Resa
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - John Nagy
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85281, USA
- Department of Life Sciences, Scottsdale Community College, Scottsdale, AZ 85256, USA
| | - Yang Kuang
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85281, USA
- Correspondence:
| |
Collapse
|
27
|
Kareva I. Different costs of therapeutic resistance in cancer: Short- and long-term impact of population heterogeneity. Math Biosci 2022; 352:108891. [PMID: 35998834 DOI: 10.1016/j.mbs.2022.108891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/28/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022]
Abstract
Therapeutic resistance continues to undercut long-term success of many promising cancer treatments. At times, development of therapeutic resistance can come at a fitness cost for the cancer cell population, which could potentially be leveraged to the patient's advantage. A mathematical formulation of such a situation was proposed by Pressley et al. (2020), who discussed two scenarios, namely, when developing therapeutic resistance can come at a cost to proliferative capacity (such as when a drug targets a growth receptor), or to the total tumor carrying capacity (such as when a drug targets neovascularization). Here we expand the analysis of the two models and evaluate both short- and long-term dynamics of a population heterogeneous with respect to resistance. We analyze the four initial distributions with respect to resistance at the time of treatment initiation: uniform, bell-shaped, exponential, and U-shaped. We show that final population composition is invariant to the initial distribution, with a single clone eventually dominating within the population; the value of the resistance parameter of the final clone depends on other system parameters but not on the initial distribution. Transitional behaviors, however, which may have more significant implications for immediate treatment decisions, depend critically on the initial distribution. Furthermore, we show that depending on the mechanism for the cost of resistance (i.e., proliferation vs carrying capacity), increase in natural cell death rate has opposite effects, with higher natural death rate selecting for less resistant cell clones in the long term for proliferation-dependent model, and selecting for more resistant cell clones for carrying capacity-dependent model, a prediction that may have implications for combination therapy with cytotoxic agents. We conclude with a discussion of strengths and limitations of using modeling for understanding treatment trajectory, as well as the promise of model-informed evolutionary steering for improved long-term therapeutic outcomes.
Collapse
Affiliation(s)
- Irina Kareva
- Department of Biomedical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
28
|
Combining Androgen Deprivation and Immunotherapy in Prostate Cancer Treatment: A Mechanistic Approach. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12146954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Due to its initial dependence on testosterone, prostate cancer patients are initially treated with androgen deprivation therapy, a form of chemical castration. However, in many cases, the cancer develops resistance to this treatment. Sipuleucel-T (Provenge), is the first live cell vaccine approved for treating patients with advanced, hormonally refractive prostate cancer. However, it has shown limited survival benefit. Recently, it has been proposed that combining Provenge with androgen deprivation may result in a better treatment outcome. Here, we develop a nonlinear dynamical systems model with a view to predicting the therapeutic potential of such a combination. Our model accounts for the mechanism of action of Provenge and the immune system response elicited by androgen deprivation. We use data from mouse xenograft experiments to calibrate and validate our model. The validated model is then used to explain the limited clinical success of Provenge, and predict optimal scheduling that maximizes the anti-tumor potential of Provenge combined with androgen deprivation. In particular, we demonstrate that the two treatments should be given concurrently, rather than sequentially, as is current practice.
Collapse
|
29
|
Angelini E, Wang Y, Zhou JX, Qian H, Huang S. A model for the intrinsic limit of cancer therapy: Duality of treatment-induced cell death and treatment-induced stemness. PLoS Comput Biol 2022; 18:e1010319. [PMID: 35877695 PMCID: PMC9352192 DOI: 10.1371/journal.pcbi.1010319] [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: 11/09/2021] [Revised: 08/04/2022] [Accepted: 06/20/2022] [Indexed: 11/23/2022] Open
Abstract
Intratumor cellular heterogeneity and non-genetic cell plasticity in tumors pose a recently recognized challenge to cancer treatment. Because of the dispersion of initial cell states within a clonal tumor cell population, a perturbation imparted by a cytocidal drug only kills a fraction of cells. Due to dynamic instability of cellular states the cells not killed are pushed by the treatment into a variety of functional states, including a "stem-like state" that confers resistance to treatment and regenerative capacity. This immanent stress-induced stemness competes against cell death in response to the same perturbation and may explain the near-inevitable recurrence after any treatment. This double-edged-sword mechanism of treatment complements the selection of preexisting resistant cells in explaining post-treatment progression. Unlike selection, the induction of a resistant state has not been systematically analyzed as an immanent cause of relapse. Here, we present a generic elementary model and analytical examination of this intrinsic limitation to therapy. We show how the relative proclivity towards cell death versus transition into a stem-like state, as a function of drug dose, establishes either a window of opportunity for containing tumors or the inevitability of progression following therapy. The model considers measurable cell behaviors independent of specific molecular pathways and provides a new theoretical framework for optimizing therapy dosing and scheduling as cancer treatment paradigms move from "maximal tolerated dose," which may promote therapy induced-stemness, to repeated "minimally effective doses" (as in adaptive therapies), which contain the tumor and avoid therapy-induced progression.
Collapse
Affiliation(s)
- Erin Angelini
- Department of Applied Mathematics, University of Washington, Seattle, Washington, United States of America
| | - Yue Wang
- Department of Applied Mathematics, University of Washington, Seattle, Washington, United States of America
- Institut des Hautes Études Scientifiques, Bures-sur-Yvette, France
| | - Joseph Xu Zhou
- Immuno-Oncology Department, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, United States of America
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Hong Qian
- Department of Applied Mathematics, University of Washington, Seattle, Washington, United States of America
| | - Sui Huang
- Institute for Systems Biology, Seattle, Washington, United States of America
| |
Collapse
|
30
|
Tang DG. Understanding and targeting prostate cancer cell heterogeneity and plasticity. Semin Cancer Biol 2022; 82:68-93. [PMID: 34844845 PMCID: PMC9106849 DOI: 10.1016/j.semcancer.2021.11.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) is a prevalent malignancy that occurs primarily in old males. Prostate tumors in different patients manifest significant inter-patient heterogeneity with respect to histo-morphological presentations and molecular architecture. An individual patient tumor also harbors genetically distinct clones in which PCa cells display intra-tumor heterogeneity in molecular features and phenotypic marker expression. This inherent PCa cell heterogeneity, e.g., in the expression of androgen receptor (AR), constitutes a barrier to the long-term therapeutic efficacy of AR-targeting therapies. Furthermore, tumor progression as well as therapeutic treatments induce PCa cell plasticity such that AR-positive PCa cells may turn into AR-negative cells and prostate tumors may switch lineage identity from adenocarcinomas to neuroendocrine-like tumors. This induced PCa cell plasticity similarly confers resistance to AR-targeting and other therapies. In this review, I first discuss PCa from the perspective of an abnormal organ development and deregulated cellular differentiation, and discuss the luminal progenitor cells as the likely cells of origin for PCa. I then focus on intrinsic PCa cell heterogeneity in treatment-naïve tumors with the presence of prostate cancer stem cells (PCSCs). I further elaborate on PCa cell plasticity induced by genetic alterations and therapeutic interventions, and present potential strategies to therapeutically tackle PCa cell heterogeneity and plasticity. My discussions will make it clear that, to achieve enduring clinical efficacy, both intrinsic PCa cell heterogeneity and induced PCa cell plasticity need to be targeted with novel combinatorial approaches.
Collapse
Affiliation(s)
- Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Experimental Therapeutics (ET) Graduate Program, The University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
31
|
Viguerie A, Grave M, Barros GF, Lorenzo G, Reali A, Coutinho A. Data-Driven Simulation of Fisher-Kolmogorov Tumor Growth Models Using Dynamic Mode Decomposition. J Biomech Eng 2022; 144:1141945. [PMID: 35771166 DOI: 10.1115/1.4054925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Indexed: 11/08/2022]
Abstract
The computer simulation of organ-scale biomechanistic models of cancer personalized via routinely collected clinical and imaging data enables to obtain patient-specific predictions of tumor growth and treatment response over the anatomy of the patient's affected organ. However, the simulation of the underlying spatiotemporal models can entail a prohibitive computational cost, which constitutes a barrier to the successful development of clinically-actionable computational technologies for personalized tumor forecasting. Here we propose to utilize Dynamic-Mode Decomposition (DMD), an unsupervised machine learning method, to construct a low-dimensional representation of cancer models and accelerate their simulation. We show that DMD may be applied to Fisher-Kolmogorov models, which constitute an established formulation to represent untreated solid tumor growth that can further accommodate other relevant cancer phenomena. Our results show that a DMD implementation of this model over a clinically-relevant parameter space can yield impressive predictions, with short to medium-term errors remaining under 1% and long-term errors remaining under 20%, despite very short training periods. In particular, we have found that, for moderate to high tumor cell diffusivity and low to moderate tumor cell proliferation rate, DMD reconstructions provide accurate, bounded-error reconstructions for all tested training periods. We posit that this data-driven approach has the potential to greatly reduce the computational overhead of personalized simulations of cancer models, thereby facilitating tumor forecasting, parameter identification, uncertainty quantification, and treatment optimization.
Collapse
Affiliation(s)
- Alex Viguerie
- Department of Mathematics, Gran Sasso Science Institute, Viale Francesco Crispi 7, L'Aquila, AQ 67100, Italy
| | - Malú Grave
- Dept. of Civil Engineering, COPPE/Federal University of Rio de Janeiro, P.O. Box 68506, RJ 21945-970, Rio de Janeiro, Brazil; Fundação Oswaldo Cruz - Fiocruz, Rua Waldemar Falcão 121, BA 40296-710, Salvador, Brazil
| | - Gabriel F Barros
- Dept. of Civil Engineering, COPPE/Federal University of Rio de Janeiro, P.O. Box 68506, RJ 21945-970, Rio de Janeiro, Brazil
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E. 24th Street, Austin, TX, 78712-1229, USA; Dipartimento di Ingegneria Civile ed Architettura, Università di Pavia, Via Ferrata 3, Pavia, PV 27100, Italy
| | - Alessandro Reali
- Dipartimento di Ingegneria Civile ed Architettura, Università di Pavia, Via Ferrata 3, Pavia, PV 27100, Italy
| | - Alvaro Coutinho
- Dept. of Civil Engineering, COPPE/Federal University of Rio de Janeiro, P.O. Box 68506, RJ 21945-970, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Bekker RA, Kim S, Pilon-Thomas S, Enderling H. Mathematical modeling of radiotherapy and its impact on tumor interactions with the immune system. Neoplasia 2022; 28:100796. [PMID: 35447601 PMCID: PMC9043662 DOI: 10.1016/j.neo.2022.100796] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/27/2022] [Accepted: 04/01/2022] [Indexed: 11/01/2022]
Abstract
Radiotherapy is a primary therapeutic modality widely utilized with curative intent. Traditionally tumor response was hypothesized to be due to high levels of cell death induced by irreparable DNA damage. However, the immunomodulatory aspect of radiation is now widely accepted. As such, interest into the combination of radiotherapy and immunotherapy is increasing, the synergy of which has the potential to improve tumor regression beyond that observed after either treatment alone. However, questions regarding the timing (sequential vs concurrent) and dose fractionation (hyper-, standard-, or hypo-fractionation) that result in improved anti-tumor immune responses, and thus potentially enhanced tumor inhibition, remain. Here we discuss the biological response to radiotherapy and its immunomodulatory properties before giving an overview of pre-clinical data and clinical trials concerned with answering these questions. Finally, we review published mathematical models of the impact of radiotherapy on tumor-immune interactions. Ranging from considering the impact of properties of the tumor microenvironment on the induction of anti-tumor responses, to the impact of choice of radiation site in the setting of metastatic disease, these models all have an underlying feature in common: the push towards personalized therapy.
Collapse
|
33
|
Wu C, Lorenzo G, Hormuth DA, Lima EABF, Slavkova KP, DiCarlo JC, Virostko J, Phillips CM, Patt D, Chung C, Yankeelov TE. Integrating mechanism-based modeling with biomedical imaging to build practical digital twins for clinical oncology. BIOPHYSICS REVIEWS 2022; 3:021304. [PMID: 35602761 PMCID: PMC9119003 DOI: 10.1063/5.0086789] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/29/2022] [Indexed: 12/11/2022]
Abstract
Digital twins employ mathematical and computational models to virtually represent a physical object (e.g., planes and human organs), predict the behavior of the object, and enable decision-making to optimize the future behavior of the object. While digital twins have been widely used in engineering for decades, their applications to oncology are only just emerging. Due to advances in experimental techniques quantitatively characterizing cancer, as well as advances in the mathematical and computational sciences, the notion of building and applying digital twins to understand tumor dynamics and personalize the care of cancer patients has been increasingly appreciated. In this review, we present the opportunities and challenges of applying digital twins in clinical oncology, with a particular focus on integrating medical imaging with mechanism-based, tissue-scale mathematical modeling. Specifically, we first introduce the general digital twin framework and then illustrate existing applications of image-guided digital twins in healthcare. Next, we detail both the imaging and modeling techniques that provide practical opportunities to build patient-specific digital twins for oncology. We then describe the current challenges and limitations in developing image-guided, mechanism-based digital twins for oncology along with potential solutions. We conclude by outlining five fundamental questions that can serve as a roadmap when designing and building a practical digital twin for oncology and attempt to provide answers for a specific application to brain cancer. We hope that this contribution provides motivation for the imaging science, oncology, and computational communities to develop practical digital twin technologies to improve the care of patients battling cancer.
Collapse
Affiliation(s)
- Chengyue Wu
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | | | - Kalina P. Slavkova
- Department of Physics, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | - Caleb M. Phillips
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Debra Patt
- Texas Oncology, Austin, Texas 78731, USA
| | - Caroline Chung
- Department of Radiation Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas 77030, USA
| | | |
Collapse
|
34
|
Thomas DS, Cisneros LH, Anderson ARA, Maley CC. In Silico Investigations of Multi-Drug Adaptive Therapy Protocols. Cancers (Basel) 2022; 14:2699. [PMID: 35681680 PMCID: PMC9179496 DOI: 10.3390/cancers14112699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
The standard of care for cancer patients aims to eradicate the tumor by killing the maximum number of cancer cells using the maximum tolerated dose (MTD) of a drug. MTD causes significant toxicity and selects for resistant cells, eventually making the tumor refractory to treatment. Adaptive therapy aims to maximize time to progression (TTP), by maintaining sensitive cells to compete with resistant cells. We explored both dose modulation (DM) protocols and fixed dose (FD) interspersed with drug holiday protocols. In contrast to previous single drug protocols, we explored the determinants of success of two-drug adaptive therapy protocols, using an agent-based model. In almost all cases, DM protocols (but not FD protocols) increased TTP relative to MTD. DM protocols worked well when there was more competition, with a higher cost of resistance, greater cell turnover, and when crowded proliferating cells could replace their neighbors. The amount that the drug dose was changed, mattered less. The more sensitive the protocol was to tumor burden changes, the better. In general, protocols that used as little drug as possible, worked best. Preclinical experiments should test these predictions, especially dose modulation protocols, with the goal of generating successful clinical trials for greater cancer control.
Collapse
Affiliation(s)
- Daniel S. Thomas
- Arizona Cancer Evolution Center, Arizona State University, Tempe, AZ 85287, USA; (D.S.T.); (L.H.C.)
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, AZ 85287, USA
| | - Luis H. Cisneros
- Arizona Cancer Evolution Center, Arizona State University, Tempe, AZ 85287, USA; (D.S.T.); (L.H.C.)
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, AZ 85287, USA
| | | | - Carlo C. Maley
- Arizona Cancer Evolution Center, Arizona State University, Tempe, AZ 85287, USA; (D.S.T.); (L.H.C.)
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, AZ 85287, USA
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ 85287, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
35
|
Spatial structure impacts adaptive therapy by shaping intra-tumoral competition. COMMUNICATIONS MEDICINE 2022; 2:46. [PMID: 35603284 PMCID: PMC9053239 DOI: 10.1038/s43856-022-00110-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/28/2022] [Indexed: 02/07/2023] Open
Abstract
Background Adaptive therapy aims to tackle cancer drug resistance by leveraging resource competition between drug-sensitive and resistant cells. Here, we present a theoretical study of intra-tumoral competition during adaptive therapy, to investigate under which circumstances it will be superior to aggressive treatment. Methods We develop and analyse a simple, 2-D, on-lattice, agent-based tumour model in which cells are classified as fully drug-sensitive or resistant. Subsequently, we compare this model to its corresponding non-spatial ordinary differential equation model, and fit it to longitudinal prostate-specific antigen data from 65 prostate cancer patients undergoing intermittent androgen deprivation therapy following biochemical recurrence. Results Leveraging the individual-based nature of our model, we explicitly demonstrate competitive suppression of resistance during adaptive therapy, and examine how different factors, such as the initial resistance fraction or resistance costs, alter competition. This not only corroborates our theoretical understanding of adaptive therapy, but also reveals that competition of resistant cells with each other may play a more important role in adaptive therapy in solid tumours than was previously thought. To conclude, we present two case studies, which demonstrate the implications of our work for: (i) mathematical modelling of adaptive therapy, and (ii) the intra-tumoral dynamics in prostate cancer patients during intermittent androgen deprivation treatment, a precursor of adaptive therapy. Conclusion Our work shows that the tumour’s spatial architecture is an important factor in adaptive therapy and provides insights into how adaptive therapy leverages both inter- and intra-specific competition to control resistance. Cancer therapy traditionally focuses on maximising tumour cell kill with the aim of achieving a cure, but such aggressive treatment can open up space for drug-resistant cells to grow. In contrast, adaptive therapy aims to leverage competition between drug-sensitive and resistant cells by adjusting treatment to maintain the tumour at a tolerable size, whilst preserving drug-sensitive cells. This approach is being tested in trials but is not yet widely used as deeper understanding of cell-cell competition is required. Here, we used a mathematical model to investigate how strongly, and with whom, resistant cells compete during continuous and adaptive therapy, and applied our insights to hormone therapy in prostate cancer where adaptive therapy has recently been successfully trialed. Our results provide new insights into how adaptive therapy works and show that, by shaping cell competition, the tumour’s spatial architecture is important in determining therapy response. Strobl et al. develop an agent-based spatial model of drug resistance in tumour cells under adaptive therapy. Using this model, they investigate how the tumour’s spatial architecture impacts intratumoural competitive dynamics of drug-sensitive vs. -resistant clones in response to therapy.
Collapse
|
36
|
Mathur D, Barnett E, Scher HI, Xavier JB. Optimizing the future: how mathematical models inform treatment schedules for cancer. Trends Cancer 2022; 8:506-516. [PMID: 35277375 DOI: 10.1016/j.trecan.2022.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/25/2022] [Accepted: 02/14/2022] [Indexed: 11/18/2022]
Abstract
For decades, mathematical models have influenced how we schedule chemotherapeutics. More recently, mathematical models have leveraged lessons from ecology, evolution, and game theory to advance predictions of optimal treatment schedules, often in a personalized medicine manner. We discuss both established and emerging therapeutic strategies that deviate from canonical standard-of-care regimens, and how mathematical models have contributed to the design of such schedules. We first examine scheduling options for single therapies and review the advantages and disadvantages of various treatment plans. We then consider the challenge of scheduling multiple therapies, and review the mathematical and clinical support for various conflicting treatment schedules. Finally, we propose how a consilience of mathematical and clinical knowledge can best determine the optimal treatment schedules for patients.
Collapse
Affiliation(s)
- Deepti Mathur
- Program for Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ethan Barnett
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joao B Xavier
- Program for Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
37
|
Howard GR, Jost TA, Yankeelov TE, Brock A. Quantification of long-term doxorubicin response dynamics in breast cancer cell lines to direct treatment schedules. PLoS Comput Biol 2022; 18:e1009104. [PMID: 35358172 PMCID: PMC9004764 DOI: 10.1371/journal.pcbi.1009104] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 04/12/2022] [Accepted: 02/07/2022] [Indexed: 01/05/2023] Open
Abstract
While acquired chemoresistance is recognized as a key challenge to treating many types of cancer, the dynamics with which drug sensitivity changes after exposure are poorly characterized. Most chemotherapeutic regimens call for repeated dosing at regular intervals, and if drug sensitivity changes on a similar time scale then the treatment interval could be optimized to improve treatment performance. Theoretical work suggests that such optimal schedules exist, but experimental confirmation has been obstructed by the difficulty of deconvolving the simultaneous processes of death, adaptation, and regrowth taking place in cancer cell populations. Here we present a method of optimizing drug schedules in vitro through iterative application of experimentally calibrated models, and demonstrate its ability to characterize dynamic changes in sensitivity to the chemotherapeutic doxorubicin in three breast cancer cell lines subjected to treatment schedules varying in concentration, interval between pulse treatments, and number of sequential pulse treatments. Cell populations are monitored longitudinally through automated imaging for 600–800 hours, and this data is used to calibrate a family of cancer growth models, each consisting of a system of ordinary differential equations, derived from the bi-exponential model which characterizes resistant and sensitive subpopulations. We identify a model incorporating both a period of growth arrest in surviving cells and a delay in the death of chemosensitive cells which outperforms the original bi-exponential growth model in Akaike Information Criterion based model selection, and use the calibrated model to quantify the performance of each drug schedule. We find that the inter-treatment interval is a key variable in determining the performance of sequential dosing schedules and identify an optimal retreatment time for each cell line which extends regrowth time by 40%-239%, demonstrating that the time scale of changes in chemosensitivity following doxorubicin exposure allows optimization of drug scheduling by varying this inter-treatment interval. Acquired chemoresistance is a common cause of treatment failure in cancer. The scheduling of a multi-dose course of chemotherapeutic treatment may influence the dynamics of acquired chemoresistance, and drug schedule optimization may increase the duration of effectiveness of a particular chemotherapeutic agent for a particular patient. Here we present a method for experimentally optimizing an in vitro drug schedule through iterative rounds of experimentation and computational analysis, and demonstrate the method’s ability to improve the performance of doxorubicin treatment in three breast carcinoma cell lines. Specifically, we find that the interval between drug exposures can be optimized while holding drug concentration and number of treatments constant, suggesting that this may be a key variable to explore in future drug schedule optimization efforts. We further use this method’s model calibration and selection process to extract information about the underlying biology of the doxorubicin response, and find that the incorporation of delays on both cell death and regrowth are necessary for accurate parameterization of cell growth data.
Collapse
Affiliation(s)
- Grant R. Howard
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Tyler A. Jost
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Thomas E. Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Oncology, The University of Texas at Austin, Austin, Texas, United States of America
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas, United States of America
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Cell and Molecular Biology, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Mathematical oncology: A new frontier in cancer biology and clinical decision making: Comment on "Improving cancer treatments via dynamical biophysical models" by M. Kuznetsov, J. Clairambault & V. Volpert. Phys Life Rev 2022; 40:60-62. [PMID: 34857476 PMCID: PMC9870030 DOI: 10.1016/j.plrev.2021.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 01/26/2023]
|
39
|
Siewe N, Friedman A. Combination therapy for mCRPC with immune checkpoint inhibitors, ADT and vaccine: A mathematical model. PLoS One 2022; 17:e0262453. [PMID: 35015785 PMCID: PMC8752026 DOI: 10.1371/journal.pone.0262453] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/23/2021] [Indexed: 11/27/2022] Open
Abstract
Metastatic castration resistant prostate cancer (mCRPC) is commonly treated by androgen deprivation therapy (ADT) in combination with chemotherapy. Immune therapy by checkpoint inhibitors, has become a powerful new tool in the treatment of melanoma and lung cancer, and it is currently being used in clinical trials in other cancers, including mCRPC. However, so far, clinical trials with PD-1 and CTLA-4 inhibitors have been disappointing. In the present paper we develop a mathematical model to assess the efficacy of any combination of ADT with cancer vaccine, PD-1 inhibitor, and CTLA-4 inhibitor. The model is represented by a system of partial differential equations (PDEs) for cells, cytokines and drugs whose density/concentration evolves in time within the tumor. Efficacy of treatment is determined by the reduction in tumor volume at the endpoint of treatment. In mice experiments with ADT and various combinations of PD-1 and CTLA-4 inhibitors, tumor volume at day 30 was always larger than the initial tumor. Our model, however, shows that we can decrease tumor volume with large enough dose; for example, with 10 fold increase in the dose of anti-PD-1, initial tumor volume will decrease by 60%. Although the treatment with ADT in combination with PD-1 inhibitor or CTLA-4 inhibitor has been disappointing in clinical trials, our simulations suggest that, disregarding negative effects, combinations of ADT with checkpoint inhibitors can be effective in reducing tumor volume if larger doses are used. This points to the need for determining the optimal combination and amounts of dose for individual patients.
Collapse
Affiliation(s)
- Nourridine Siewe
- School of Mathematical Sciences, College of Science, Rochester Institute of Technology, Rochester, New York, United States of America
| | - Avner Friedman
- Mathematical Biosciences Institute & Department of Mathematics, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
40
|
Pasetto S, Enderling H, Gatenby RA, Brady-Nicholls R. Intermittent Hormone Therapy Models Analysis and Bayesian Model Comparison for Prostate Cancer. Bull Math Biol 2021; 84:2. [PMID: 34797430 PMCID: PMC8604892 DOI: 10.1007/s11538-021-00953-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 09/27/2021] [Indexed: 11/26/2022]
Abstract
The prostate is an exocrine gland of the male reproductive system dependent on androgens (testosterone and dihydrotestosterone) for development and maintenance. First-line therapy for prostate cancer includes androgen deprivation therapy (ADT), depriving both the normal and malignant prostate cells of androgens required for proliferation and survival. A significant problem with continuous ADT at the maximum tolerable dose is the insurgence of cancer cell resistance. In recent years, intermittent ADT has been proposed as an alternative to continuous ADT, limiting toxicities and delaying time-to-progression. Several mathematical models with different biological resistance mechanisms have been considered to simulate intermittent ADT response dynamics. We present a comparison between 13 of these intermittent dynamical models and assess their ability to describe prostate-specific antigen (PSA) dynamics. The models are calibrated to longitudinal PSA data from the Canadian Prospective Phase II Trial of intermittent ADT for locally advanced prostate cancer. We perform Bayesian inference and model analysis over the models’ space of parameters on- and off-treatment to determine each model’s strength and weakness in describing the patient-specific PSA dynamics. Additionally, we carry out a classical Bayesian model comparison on the models’ evidence to determine the models with the highest likelihood to simulate the clinically observed dynamics. Our analysis identifies several models with critical abilities to disentangle between relapsing and not relapsing patients, together with parameter intervals where the critical points’ basin of attraction might be exploited for clinical purposes. Finally, within the Bayesian model comparison framework, we identify the most compelling models in the description of the clinical data.
Collapse
Affiliation(s)
- S Pasetto
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| | - H Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.,Department of Radiation Oncology, H. Lee Moffitt Cancer and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.,Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - R A Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.,Department of Radiology, H. Lee Moffitt Cancer and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - R Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
41
|
Yang J, Xu Z, Wu WKK, Chu Q, Zhang Q. GraphSynergy: a network-inspired deep learning model for anticancer drug combination prediction. J Am Med Inform Assoc 2021; 28:2336-2345. [PMID: 34472609 PMCID: PMC8510276 DOI: 10.1093/jamia/ocab162] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/15/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To develop an end-to-end deep learning framework based on a protein-protein interaction (PPI) network to make synergistic anticancer drug combination predictions. MATERIALS AND METHODS We propose a deep learning framework named Graph Convolutional Network for Drug Synergy (GraphSynergy). GraphSynergy adapts a spatial-based Graph Convolutional Network component to encode the high-order topological relationships in the PPI network of protein modules targeted by a pair of drugs, as well as the protein modules associated with a specific cancer cell line. The pharmacological effects of drug combinations are explicitly evaluated by their therapy and toxicity scores. An attention component is also introduced in GraphSynergy, which aims to capture the pivotal proteins that play a part in both PPI network and biomolecular interactions between drug combinations and cancer cell lines. RESULTS GraphSynergy outperforms the classic and state-of-the-art models in predicting synergistic drug combinations on the 2 latest drug combination datasets. Specifically, GraphSynergy achieves accuracy values of 0.7553 (11.94% improvement compared to DeepSynergy, the latest published drug combination prediction algorithm) and 0.7557 (10.95% improvement compared to DeepSynergy) on DrugCombDB and Oncology-Screen datasets, respectively. Furthermore, the proteins allocated with high contribution weights during the training of GraphSynergy are proved to play a role in view of molecular functions and biological processes, such as transcription and transcription regulation. CONCLUSION The introduction of topological relations between drug combination and cell line within the PPI network can significantly improve the capability of synergistic drug combination identification.
Collapse
Affiliation(s)
- Jiannan Yang
- School of Data Science, City University of Hong Kong, Hong Kong,
S.A.R. of China
| | - Zhongzhi Xu
- Hong Kong Jockey Club Centre for Suicide Research and Prevention, The
University of Hong Kong, Hong Kong, S.A.R. of China
| | - William Ka Kei Wu
- Department of Anaesthesia and Intensive Care, Chinese University of Hong
Kong, Hong Kong, S.A.R. of China
| | - Qian Chu
- Department of Thoracic Oncology, Tongji Hospital, Huazhong University of
Science and Technology, Wuhan, China
| | - Qingpeng Zhang
- School of Data Science, City University of Hong Kong, Hong Kong,
S.A.R. of China
| |
Collapse
|
42
|
Intermittent radiotherapy as alternative treatment for recurrent high grade glioma: a modeling study based on longitudinal tumor measurements. Sci Rep 2021; 11:20219. [PMID: 34642366 PMCID: PMC8511136 DOI: 10.1038/s41598-021-99507-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
Recurrent high grade glioma patients face a poor prognosis for which no curative treatment option currently exists. In contrast to prescribing high dose hypofractionated stereotactic radiotherapy (HFSRT, [Formula: see text] Gy [Formula: see text] 5 in daily fractions) with debulking intent, we suggest a personalized treatment strategy to improve tumor control by delivering high dose intermittent radiation treatment (iRT, [Formula: see text] Gy [Formula: see text] 1 every 6 weeks). We performed a simulation analysis to compare HFSRT, iRT and iRT plus boost ([Formula: see text] Gy [Formula: see text] 3 in daily fractions at time of progression) based on a mathematical model of tumor growth, radiation response and patient-specific evolution of resistance to additional treatments (pembrolizumab and bevacizumab). Model parameters were fitted from tumor growth curves of 16 patients enrolled in the phase 1 NCT02313272 trial that combined HFSRT with bevacizumab and pembrolizumab. Then, iRT +/- boost treatments were simulated and compared to HFSRT based on time to tumor regrowth. The modeling results demonstrated that iRT + boost(- boost) treatment was equal or superior to HFSRT in 15(11) out of 16 cases and that patients that remained responsive to pembrolizumab and bevacizumab would benefit most from iRT. Time to progression could be prolonged through the application of additional, intermittently delivered fractions. iRT hence provides a promising treatment option for recurrent high grade glioma patients for prospective clinical evaluation.
Collapse
|
43
|
Targeting Cellular DNA Damage Responses in Cancer: An In Vitro-Calibrated Agent-Based Model Simulating Monolayer and Spheroid Treatment Responses to ATR-Inhibiting Drugs. Bull Math Biol 2021; 83:103. [PMID: 34459993 PMCID: PMC8405495 DOI: 10.1007/s11538-021-00935-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/10/2021] [Indexed: 11/26/2022]
Abstract
We combine a systems pharmacology approach with an agent-based modelling approach to simulate LoVo cells subjected to AZD6738, an ATR (ataxia–telangiectasia-mutated and rad3-related kinase) inhibiting anti-cancer drug that can hinder tumour proliferation by targeting cellular DNA damage responses. The agent-based model used in this study is governed by a set of empirically observable rules. By adjusting only the rules when moving between monolayer and multi-cellular tumour spheroid simulations, whilst keeping the fundamental mathematical model and parameters intact, the agent-based model is first parameterised by monolayer in vitro data and is thereafter used to simulate treatment responses in in vitro tumour spheroids subjected to dynamic drug delivery. Spheroid simulations are subsequently compared to in vivo data from xenografts in mice. The spheroid simulations are able to capture the dynamics of in vivo tumour growth and regression for approximately 8 days post-tumour injection. Translating quantitative information between in vitro and in vivo research remains a scientifically and financially challenging step in preclinical drug development processes. However, well-developed in silico tools can be used to facilitate this in vitro to in vivo translation, and in this article, we exemplify how data-driven, agent-based models can be used to bridge the gap between in vitro and in vivo research. We further highlight how agent-based models, that are currently underutilised in pharmaceutical contexts, can be used in preclinical drug development.
Collapse
|
44
|
Brady-Nicholls R, Zhang J, Zhang T, Wang AZ, Butler R, Gatenby RA, Enderling H. Predicting patient-specific response to adaptive therapy in metastatic castration-resistant prostate cancer using prostate-specific antigen dynamics. Neoplasia 2021; 23:851-858. [PMID: 34298234 PMCID: PMC8322456 DOI: 10.1016/j.neo.2021.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 11/27/2022] Open
Abstract
Abiraterone acetate (AA) has been proven effective for metastatic castration-resistant prostate cancer (mCRPC), and it has been proposed that adaptive AA may reduce toxicity and prolong time to progression, when compared to continuous AA. We developed a simple quantitative model of prostate-specific antigen (PSA) dynamics to evaluate prostate cancer (PCa) stem cell enrichment as a plausible driver of AA treatment resistance. The model incorporated PCa stem cells, non-stem PCa cells and PSA dynamics during adaptive therapy. A leave-one-out analysis was used to calibrate and validate the model against longitudinal PSA data from 16 mCRPC patients receiving adaptive AA in a pilot clinical study. Early PSA treatment response dynamics were used to predict patient response to subsequent treatment. We extended the model to incorporate metastatic burden and also investigated the survival benefit of adding concurrent chemotherapy for patients predicted to become resistant. Model simulations demonstrated PCa stem cell self-renewal as a plausible driver of resistance to adaptive therapy. Evolutionary dynamics from individual treatment cycles combined with metastatic burden measurements predicted patient response with 81% accuracy (specificity=92%, sensitivity=50%). In those patients predicted to progress, simulations of the addition of concurrent chemotherapy suggest a benefit between 1% and 11% reduction in probability of progression when compared to adaptive AA alone. This study developed the first mCRPC patient-specific mathematical model to use early PSA treatment response dynamics to predict subsequent responses to adaptive AA, demonstrating the putative value of integrating mathematical modeling into clinical decision making.
Collapse
Affiliation(s)
- Renee Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, 20 Duke Medicine Cir, Durham, NC, USA
| | - Andrew Z Wang
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robert Butler
- Physical Sciences Oncology Center, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, , USA
| | - Robert A Gatenby
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
45
|
Chen S, Wang C, Zhu R, Zhu S, Zhang G. Predicting prognosis in acute myeloid leukemia patients by surface-enhanced Raman spectroscopy. Nanomedicine (Lond) 2021; 16:1873-1885. [PMID: 34269596 DOI: 10.2217/nnm-2021-0199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: To develop a timely and accurate method for predicting acute myeloid leukemia (AML) prognosis after chemotherapy treatment by surface-enhanced Raman spectroscopy (SERS). Methods: Biomolecular differences between AML patients with good and poor prognosis and individuals without AML were investigated based on SERS measurements of bone marrow supernatant fluid samples. Multivariate analysis was implemented on the SERS measurements to establish an AML prognostic model. Results: Significant differences in amino acid, saccharide and lipid levels were observed between AML patients with good and poor prognoses. The AML prognostic model achieved a prediction accuracy of 84.78%. Conclusion: The proposed method could be a potential diagnostic tool for timely and precise prediction of AML prognosis.
Collapse
Affiliation(s)
- Shuo Chen
- College of Medicine & Biological Information Engineering, Northeastern University, No. 500 Wisdom Street, Shenyang, 110169, China.,Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, No. 500 Wisdom Street, Shenyang, 110169, China
| | - Chunmeng Wang
- College of Medicine & Biological Information Engineering, Northeastern University, No. 500 Wisdom Street, Shenyang, 110169, China
| | - Ruochen Zhu
- College of Medicine & Biological Information Engineering, Northeastern University, No. 500 Wisdom Street, Shenyang, 110169, China
| | - Shanshan Zhu
- Research Institute for Medical & Biological Engineering, Ningbo University, No. 818 Fenghua Road, Ningbo, 315211, China
| | - Guojun Zhang
- Department of Hematology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110022, China
| |
Collapse
|
46
|
Jain HV, Sorribes IC, Handelman SK, Barnaby J, Jackson TL. Standing Variations Modeling Captures Inter-Individual Heterogeneity in a Deterministic Model of Prostate Cancer Response to Combination Therapy. Cancers (Basel) 2021; 13:1872. [PMID: 33919753 PMCID: PMC8070719 DOI: 10.3390/cancers13081872] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 12/29/2022] Open
Abstract
Sipuleucel-T (Provenge) is the first live cell vaccine approved for advanced, hormonally refractive prostate cancer. However, survival benefit is modest and the optimal combination or schedule of sipuleucel-T with androgen depletion remains unknown. We employ a nonlinear dynamical systems approach to modeling the response of hormonally refractive prostate cancer to sipuleucel-T. Our mechanistic model incorporates the immune response to the cancer elicited by vaccination, and the effect of androgen depletion therapy. Because only a fraction of patients benefit from sipuleucel-T treatment, inter-individual heterogeneity is clearly crucial. Therefore, we introduce our novel approach, Standing Variations Modeling, which exploits inestimability of model parameters to capture heterogeneity in a deterministic model. We use data from mouse xenograft experiments to infer distributions on parameters critical to tumor growth and to the resultant immune response. Sampling model parameters from these distributions allows us to represent heterogeneity, both at the level of the tumor cells and the individual (mouse) being treated. Our model simulations explain the limited success of sipuleucel-T observed in practice, and predict an optimal combination regime that maximizes predicted efficacy. This approach will generalize to a range of emerging cancer immunotherapies.
Collapse
Affiliation(s)
- Harsh Vardhan Jain
- Department of Mathematics & Statistics, University of Minnesota Duluth, Duluth, MN 55812, USA
| | | | - Samuel K. Handelman
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Johnna Barnaby
- Department of Mathematics, Shippensburg University, Shippensburg, PA 17257, USA;
| | | |
Collapse
|
47
|
Kimmel GJ, Locke FL, Altrock PM. The roles of T cell competition and stochastic extinction events in chimeric antigen receptor T cell therapy. Proc Biol Sci 2021; 288:20210229. [PMID: 33757357 PMCID: PMC8059581 DOI: 10.1098/rspb.2021.0229] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a remarkably effective immunotherapy that relies on in vivo expansion of engineered CAR T cells, after lymphodepletion (LD) by chemotherapy. The quantitative laws underlying this expansion and subsequent tumour eradication remain unknown. We develop a mathematical model of T cell-tumour cell interactions and demonstrate that expansion can be explained by immune reconstitution dynamics after LD and competition among T cells. CAR T cells rapidly grow and engage tumour cells but experience an emerging growth rate disadvantage compared to normal T cells. Since tumour eradication is deterministically unstable in our model, we define cure as a stochastic event, which, even when likely, can occur at variable times. However, we show that variability in timing is largely determined by patient variability. While cure events impacted by these fluctuations occur early and are narrowly distributed, progression events occur late and are more widely distributed in time. We parameterized our model using population-level CAR T cell and tumour data over time and compare our predictions with progression-free survival rates. We find that therapy could be improved by optimizing the tumour-killing rate and the CAR T cells' ability to adapt, as quantified by their carrying capacity. Our tumour extinction model can be leveraged to examine why therapy works in some patients but not others, and to better understand the interplay of deterministic and stochastic effects on outcomes. For example, our model implies that LD before a second CAR T injection is necessary.
Collapse
Affiliation(s)
- Gregory J Kimmel
- Department of Integrated Mathematical Oncology, Research Institute, Tampa, FL, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Research Institute, Tampa, FL, USA
| | - Philipp M Altrock
- Department of Integrated Mathematical Oncology, Research Institute, Tampa, FL, USA.,Department of Blood and Marrow Transplant and Cellular Immunotherapy, Research Institute, Tampa, FL, USA.,Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
48
|
Barnaby JP, Sorribes IC, Jain HV. Relating prostate‐specific antigen leakage with vascular tumor growth in a mathematical model of prostate cancer response to androgen deprivation. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021. [DOI: 10.1002/cso2.1014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Johnna P. Barnaby
- Department of Mathematics Shippensberg University Shippensburg Pennsylvania USA
| | | | - Harsh Vardhan Jain
- Department of Mathematics and Statistics University of Minnesota Duluth Duluth Minnesota USA
| |
Collapse
|
49
|
A Quantitative Paradigm for Decision-Making in Precision Oncology. Trends Cancer 2021; 7:293-300. [PMID: 33637444 DOI: 10.1016/j.trecan.2021.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 11/24/2022]
Abstract
The complexity and variability of cancer progression necessitate a quantitative paradigm for therapeutic decision-making that is dynamic, personalized, and capable of identifying optimal treatment strategies for individual patients under substantial uncertainty. Here, we discuss the core components and challenges of such an approach and highlight the need for comprehensive longitudinal clinical and molecular data integration in its development. We describe the complementary and varied roles of mathematical modeling and machine learning in constructing dynamic optimal cancer treatment strategies and highlight the potential of reinforcement learning approaches in this endeavor.
Collapse
|
50
|
Affiliation(s)
- Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33647, USA
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, 18051 Rostock, Germany
| |
Collapse
|