ONCO-01
Afton Widdershins
Pennsylvania State University College of Medicine
Poster ID: ONCO-01 (Session: PS01)
"Exploring Impact of Treatment Design on Ability to Leverage Intratumor Competition and Control Multiply Resistant Populations."
BACKGROUND: Cancer is a disease with an incredible ability to adapt when exposed to clinical treatment. This evolution of resistance presents a real challenge to the long-term success of treatments like targeted therapies. Taking into account tumor evolutionary dynamics like inter-clonal competition could provide insight to how to design therapies to best utilize the drugs that are already available to clinicians.
METHODS: To allow for a model that could be validated through laboratory work, we worked with an ordinary differential equation (ODE) system of an in vitro cell population approximating a heterogeneous tumor. The ODE system is composed of four individual cell populations that respond to two drugs, with one cell population being fully susceptible, one being fully resistant, and the other two populations being resistant to one or the other drug. Competition and growth are modeled through the logistic growth term, while cell death is based on each drug’s concentration. Three different regimen categories were simulated using MATLAB – alternation, combination, and sequential. In order to explore fully explore different regimen designs, drug concentration was varied in all of the regimens, while the ratio between the two drugs was varied in the combination regimen settings and the frequency of alternation was varied in the alternation settings. The population parameters of total initial cell burden and the ratios between susceptible and various resistant populations were also varied to explore patient impact on regimen effectiveness. A regimen’s ability to control a population was defined as how long it could maintain the population below a chosen threshold less than the carrying capacity of the system. The regimens were then analyzed for their ability to control both the fully resistant population and the total population and compared to the control achieved by minimal and maximal competition scenarios established by previous work.
RESULTS: The most important parameter varied in regimen design was concentration, as alternation and combination regimens with the same total concentration achieved grossly similar population control. In terms of the impact of alternation frequency, daily and weekly alternation had similar control of the fully resistant population and total population. Longer frequency alternations, like monthly, achieved better fully resistant control but had worse total population control.
DISCUSSION: Earlier work suggests that incorporating competition into regimen design could extend control of a tumor population, though the similarities between the performance of different regimens suggests that maintaining a certain level of competition is more important than the method used to manage the population. However, this resemblance may be attributable to the simplicity of the model and the lack of consideration of consequences like toxicity for each regimen or of tumor abilities like mutation. Future work would include analysis to see if inclusion of more complex pharmacokinetics or more complex cell behaviors significantly change these results.
ONCO-02
Alejandro Bertolet
Massachusetts General Hospital and Harvard Medical School
Poster ID: ONCO-02 (Session: PS01)
"The Microdosimetric Gamma Model: A Novel Approach to Predict Analytically DNA Damage Based on In-Silico Simulations"
Purpose: Quantifying and characterizing DNA damage is critical for optimizing radiation therapy treatments, particularly in advanced modalities like proton and alpha-targeted therapy. This study presents the Microdosimetric Gamma Model (MGM), a novel approach that predicts DNA damage properties by utilizing microdosimetry theory.
Methods: MGM provides the number of DNA damage sites and their complexities, which follow a Gamma distribution. Unlike current methods, MGM can characterize DNA damage for beams with multi-energy components, various time configurations, and spatial distributions. The output can be incorporated into repair models to predict cell killing, protein recruitment, chromosome aberrations, and other biological effects. We validated the MGM using TOPAS-nBio simulations for various radiation types.
Results: MGM demonstrated excellent agreement with the simulated data, accurately predicting damage complexities for protons and alpha particles. We also predicted survival fraction curves for different cell lines, providing insights into the relative biological effectiveness (RBE) of different radiation types.
Conclusions: The Microdosimetric Gamma Model offers a flexible framework for studying ionizing radiation's energy, time, and spatial aspects. It is a valuable tool for understanding and optimizing the biological effects of radiation therapy modalities like proton therapy, targeted alpha therapy, and helium therapy.
ONCO-03
Elmar Bucher
Indiana University
Poster ID: ONCO-03 (Session: PS01)
"Agent-based Modeling of Multi-compartment Tumor Organoid Utilizing the PhysiCell Software Framework"
Tumor cell line organoid cultures are widely used in wet lab cancer research. Different from monolayer cell cultures, organoids preserve many phenotypic features demonstrated by cancer cells in vivo. In contrast to tissue microarrays, organoids offer a simplified, controllable environment. Recently, two-compartment matrigel/collagen1 organoids were developed [Lee2022], enabling scientists to mimic DCIS (ductal carcinoma in situ), IDS (invasive ductal carcinoma), and PDAC (pancreatic ductal adenocarcinoma) in extracellular matrix environments, as well as healthy mammary epithelial and fallopian tube epithelial extracellular matrix systems.
Utilizing the C++ based PhysiCell software framework [Ghaffarizade2018], we implemented an agent-based model of these two-compartment organoids. Our model was calibrated on the available data from a study from Crawford et al., who used these two-compartment organoids to explore the effect of collagen1 density conditions on cancer cell proliferation and invasion ability. Based on the results, the authors suggested that the cancer cell’s proliferation and invasiveness are being linked to cell-extracellular matrix friction [Crawford2022]. In our research work, we determine whether we can recapitulate these wet lab experiment findings.
Having a mathematical model which can capture the emergent phenomena, will extend our understanding of the two-compartment organoid wet lab model. Furthermore, the mathematical model makes it possible to quickly process a variety of experimental parameter settings in silico. The results will help to determine and plan the most interesting experimental parameters to explore in the wet lab.
[Lee2022] https://doi.org/10.1016/j.mattod.2022.07.006 [Crawford2022] https://doi.org/10.1101/2022.11.15.516548 [Ghaffarizade2018] https://doi.org/10.1371/journal.pcbi.1005991
ONCO-04
Erin Angelini
University of Washington
Poster ID: ONCO-04 (Session: PS01)
"A model for the intrinsic limit of cancer therapy: Duality of treatment-induced cell death and treatment-induced stemness"
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.
ONCO-05
Gbocho Masato Terasaki
University of California, Merced
Poster ID: ONCO-05 (Session: PS01)
"Merging Traditional Scientific Computing with Data Science to Develop a New Prediction Engine for Brain Cancer"
Glioblastoma multiforme (GBM) is one of the fastest-growing brain tumors and it has very low survival rates. Mathematical modeling can be used to predict the growth and treatment of brain cancer. However, one of the difficulties lies in the ability to estimate patient-specific parameters in the mathematical model from magnetic resonance imaging (MRI) data. We constructed a numerical solver to simulate tumor growth over a realistic 3D brain geometry derived from segmented-MRI. Then, using information about the size of the different glioma sub-regions, we are developing a method that estimates the patient-specific model parameters to inform the forward simulation. Ultimately, we hope to predict the overall survival of a patient from a single pre- operative scan.
ONCO-06
Javier C. Urcuyo Acevedo
Case Western Reserve University
Poster ID: ONCO-06 (Session: PS01)
"Exploring tumor evolution under the influence of the immune system"
While tumoral heterogeneity play a major role in the development of malignancy, the tumor microenvironment and the initial immune response are equally as important. The immune system is responsible for cancer surveillance and the initial suppression of malignancy. However, with the correct evolutionary mutations, cancer immunoediting can result in immune evasion. Yet, few models incorporate immune components to study the progression and impact of such mutations. In this work, we developed an agent-based model to explore this fitness strategy represented as an evolutionary game. Then, in a phased experimental approach, we plan to validate our model with various immune components, such as CD8+ T cells, NK cells, and other lymphocytes. Ultimately, this work will begin to elucidate the impact of the immune system on cancer evolution and allow us to begin to steer evolution towards more favorable outcomes.
ONCO-07
John Metzcar
Indiana University
Poster ID: ONCO-07 (Session: PS01)
"Using multiscale simulations to assess solutions to the Boolean network target control problem"
Boolean networks, or logical models, are proven methods for simulating a cell’s response to its environment [1], [2]. In these models, nodes represent components of the system, such as genes or proteins, and an edge from a “parent” node to a “child” node indicates that the parent has a causal influence on the child, such as a transcription factor activating a gene. Each node is assigned a time-varying binary variable that can be ON (representing presence or activity of the system component) or OFF (representing its absence or inactivity). Often, a node state (or set of node states) is taken to represent a cellular behavior of interest. In the context of Boolean networks, the process of identifying interventions that lead to a particular cellular behavior (encoded as node states that represent known phenotypic markers) is called the target control problem [3], [4]. We address this problem in selected cancer-related Boolean networks by developing and applying a new method for edgetic perturbations, which involves intervening in specific interactions (e.g., analogous to blocking specific binding sites) rather than suppressing entire biomolecules. We separately determine node interventions solving the target control problem using two previously published methods: one based on a mean field approximation and a second, percolation-based method [5], [6]. We then implement the identified node and edge interventions in a multiscale context using the combined agent-based and Boolean network simulator PhysiBoSS [7], [8]. Using this approach, we test the effectiveness of the interventions devised in an isolated, single-cell context in a more realistic in silico environment that can account for spatial features (such as chemical gradients), heterogeneity of signal response in cell populations, and cell-cell interactions.
[1] A. A. Hemedan, A. Niarakis, R. Schneider, and M. Ostaszewski, “Boolean modelling as a logic-based dynamic approach in systems medicine,” Computational and Structural Biotechnology Journal, vol. 20, pp. 3161–3172, Jan. 2022, doi: 10.1016/j.csbj.2022.06.035. [2] J. D. Schwab, S. D. Kühlwein, N. Ikonomi, M. Kühl, and H. A. Kestler, “Concepts in Boolean network modeling: What do they all mean?,” Computational and Structural Biotechnology Journal, vol. 18, pp. 571–582, 2020, doi: 10.1016/j.csbj.2020.03.001. [3] J. C. Rozum, D. Deritei, K. H. Park, J. Gómez Tejeda Zañudo, and R. Albert, “pystablemotifs: Python library for attractor identification and control in Boolean networks,” Bioinformatics, vol. 38, no. 5, pp. 1465–1466, Mar. 2022, doi: 10.1093/bioinformatics/btab825. [4] C. Su and J. Pang, “A Dynamics-based Approach for the Target Control of Boolean Networks,” in Proceedings of the 11th ACM International Conference on Bioinformatics, Computational Biology and Health Informatics, in BCB ’20. New York, NY, USA: Association for Computing Machinery, Nov. 2020, pp. 1–8. doi: 10.1145/3388440.3412464. [5] T. Parmer, L. M. Rocha, and F. Radicchi, “Influence maximization in Boolean networks,” Nature Communications, vol. 13, no. 1, p. 3457, Jun. 2022, doi: 10.1038/s41467-022-31066-0. [6] J. G. T. Zañudo and R. Albert, “Cell Fate Reprogramming by Control of Intracellular Network Dynamics,” PLOS Computational Biology, vol. 11, no. 4, p. e1004193, Apr. 2015, doi: 10.1371/journal.pcbi.1004193. [7] G. Letort et al., “PhysiBoSS: a multi-scale agent-based modelling framework integrating physical dimension and cell signalling,” Bioinformatics, vol. 35, no. 7, pp. 1188–1196, Apr. 2019, doi: 10.1093/bioinformatics/bty766. [8] M. Ponce-de-Leon et al., “PhysiBoSS 2.0: a sustainable integration of stochastic Boolean and agent-based modelling frameworks.” bioRxiv, p. 2022.01.06.468363, Mar. 27, 2023. doi: 10.1101/2022.01.06.468363.
ONCO-08
Lee Curtin
Mayo Clinic
Poster ID: ONCO-08 (Session: PS01)
"Transcriptomic Analysis of Image-Localized High-Grade Glioma Biopsies Reveals Meaningful Cellular States"
High-grade glioma continues to have dismal survival with current standard-of-care treatment, owing in part to its intra- and inter-patient heterogeneity. Typical diagnostic clinical biopsies are taken from the dense tumor core to determine the presence of abnormal cells and the status of a few key genes. However, the tumor core is removed during surgery, leaving behind possibly genetically, transcriptomically and/or phenotypically distinct invasive margins that repopulate the disease. As these remaining populations are the ones ultimately being treated, it is important to know their compositional differences from the tumor core. We aim to identify the phenotypic niches defined by the relative composition of key cellular populations and understand their variation amongst patients. We have established an image-localized research biopsy study, that samples from both the invasive margin and tumor core. From this protocol, we currently have 202 samples from 58 patients with available bulk RNA-Seq, collected between Mayo Clinic and Barrow Neurological Institute. Using a single-cell reference dataset from our collaborators at Columbia University, we used CIBERSORTx, a support vector machine deconvolution method, to predict relative abundances of normal, glioma, and immune cell states for each sample. We also applied Monocle, an algorithm that uses reversed graph embedding, to this dataset. Monocle orders samples on a low dimensional space by pseudotime, and provides a graph of transitions between end states. We find that these cell state abundances connect to patient survival and show regional differences. We analyze the robustness of these methods, and highlight the importance of characterizing residual glioma to better understand the recurrent disease.
ONCO-09
Malgorzata Tyczynska Weh
Moffitt Cancer Center
Poster ID: ONCO-09 (Session: PS01)
"Modeling selection for evolvability in the evolution of cancer therapy resistance"
Despite rapid initial responses and low toxicity, targeted therapies commonly fail to provide long-term benefits to cancer patients due to the development of therapy resistance. In multiple solid tumors, this resistance emerges due to gradual, multifactorial adaptation, i.e., a selective process combining genetic and non-genetic methods of cell diversification. This suggests a significant link between the evolution of cancer treatment resistance and evolvability – a selective trait of generating heritable phenotypic variation. However, the interplay between selection, evolvability, and resistance has not yet been fully investigated. We addressed this problem by studying the selection for mutator phenotype.
The mutator phenotype is common in many cancers and results from errors in DNA repair mechanisms. This phenotype generates mutations at a higher frequency than other phenotypes. Since mutations can both benefit or reduce cell viability, we hypothesized that the selection for a mutator phenotype changes during the evolution of resistance to cancer targeted therapies.
We tested this hypothesis by developing a 2D on-lattice Agent-Based Model (ABM). In the model, a cell can die, divide and mutate, yet mutations have a stochastic impact that can be beneficial, neutral, or deleterious for the individual cell fitness. Consequently, the resistance emerges as a stochastic event depending on the mutation frequency. Our results demonstrate that 1) the mutator phenotype initially accelerates adaptation to treatment, but 2) only intermediate mutation frequencies can sustain high fitness long-term.
This work provides a versatile experimental platform that can be adjusted to study the evolution of resistance in other cancers and treatments. Moreover, our results challenge the commonly held assumption that resistance develops only due to pre-existing driver mutations and provide an opportunity to integrate evolutionary theory and oncology to improve treatment in cancer patients.
ONCO-10
Maximilian Strobl
Cleveland Clinic
Poster ID: ONCO-10 (Session: PS01)
"Using mathematical modeling to design protocols for preclinical testing of evolutionary therapies"
Over the past two decades it has become clear that cancers are complex and evolving diseases. Genetic and non-genetic processes generate diverse subpopulations of tumor cells which can thrive under a variety of conditions and stressors. This provides a rich pool of variation that by means of natural selection and continued evolution enables adaptation to even the most modern treatments, especially in advanced cancers. Based on this novel understanding, so-called “evolutionary therapy” or “evolution-informed treatment strategies” have emerged which try to leverage, and potentially even steer, tumour evolution by strategically and dynamically sequencing and combining existing therapies and adjusting dose levels. In particular, adaptive therapy, which dynamically changes treatment levels to maintain drug-sensitive cells in order to competitively suppress emerging drug resistance, has produced a number of promising theoretical, preclinical and also clinical results. However, unlike for new drugs for which there are clear established frameworks for translation from bench to bedside, the design of preclinical protocols to ensure efficacy and safety of evolutionary therapies is an open question.
In this study, we use mathematical modeling to develop and interpret in vitro experimental protocols and apply them towards the development of an adaptive therapy for Osimertinib for the treatment of Non-Small Cell Lung Cancer. In the first step, we consider the question of how to measure ecological interactions between tumor subpopulations. To do so, we build on the “Game Assay” previously developed by our group, in which cells are co-cultured at different frequencies to measure how a population’s fitness depends on its frequency in the environment. Using an agent-based model of our in vitro experiments, we study how different aspects of the design (number of replicates, number of proportions) and analysis (regression technique, regression window) impact the accuracy and precision of the assay. Subsequently, we use our optimized protocol to quantify the frequency-dependent interactions between Osimertinib sensitive and resistant PC9 cells under different drug levels. In the next step, we use our model to explore whether and how so-obtained fitness differences translate to the ability to steer the composition of the tumor in long-term in vitro experiments, in which cells are co-cultured and passaged at regular intervals. In particular, we explore the role of the population size, passaging frequency, and passage fraction (proportion of cells carried forward to next passage). To conclude, we will present preliminary data in which we use this assay to trial a potential adaptive Osimertinib therapy protocol in vitro. Overall, we demonstrate how mathematical models can help to understand and improve experimental assays, and we contribute towards the important discussion as to how to translate evolutionary therapies from the blackboard to the bedside.
ONCO-11
Natalie Meacham
University of California, Merced
Poster ID: ONCO-11 (Session: PS01)
"An Inverse Problem to Recover Sensitivity to Treatment in Prostate Cancer Tumors"
Resistance to prostate cancer treatment is a serious concern in modern oncology due to the risk it poses for poor patient outcomes. A key facet of treatment resistance is that traditional therapies can select for resistant cells. Understanding the heterogeneity of sensitivity to treatment in heterogeneous tumors is key to predicting and delaying the time to treatment resistance. We construct a novel random differential equation (RDE) model that incorporates sensitivity to treatment, then use inverse problem methods to recover the distribution of sensitive and resistant cells from noisy simulated data. We use the Akaike Information Criteria (AIC) to pinpoint the optimal mesh for the recovered distribution, which can help optimize individual treatment plans.
ONCO-12
Nicholas Harbour
The University of Nottingham
Poster ID: ONCO-12 (Session: PS01)
"Mathematical modelling of interacting sub-populations in glioblastoma"
One of the major challenges in successfully treating glioblastoma (GBM) is the significant heterogeneity in cellular composition observed within and between patients. Recent single cell transcriptomics suggests there can be as many as eighteen distinct cell types in a single tumour [1]. Furthermore, advances in cellular deconvolution techniques, such as CIBERSORTx, allow us to accurately determine the cellular composition of imaged localised biopsies from bulk RNA-Seq [2]. Understanding this heterogeneity and how the complex interactions between cellular populations impacts the progression of GBM may lead to novel treatments which exploit the unique cellular composition within individual tumours. We group these eighteen cell types into sub-populations, e.g., glioma, immune, astrocyte, then attempt to learn the dynamics of these sub-populations by considering various interacting ODE/PDE models. Typically, a GBM patient will have biopsies taken at most twice, as well as only a handful of MRI scans. Therefore, the number of temporal data points to fit any model to are very limited. Thus, we apply trajectory inference methods, such as Monocle, to biopsy data, which allows us to order samples via pseudotime, an arbitrary unit of progress akin to real time [3]. We illustrate our modelling approach with a simplified two species Lotka-Volterra style competition model.
[1] O. Al-Dalahmah, et al., Re-convolving the compositional landscape of primary and recurrent glioblastoma using single nucleus RNA sequencing. bioRxiv (2021) https://doi.org/10.1101/2021.07.06.451295
[2] C. B. Steen, C. L. Liu, A. A. Alizadeh, A. M. Newman, Profiling Cell Type Abundance and Expression in Bulk Tissues with CIBERSORTx. Methods Mol. Biol. 2117, 135–157 (2020).
[3] C. Trapnell, et al., The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).
ONCO-13
Temitope O. Benson
University at Buffalo, The State University of New York, Buffalo, NY
Poster ID: ONCO-13 (Session: PS01)
"A Computational Model of Metastatic Cancer Cell Migration Phenotype: Single and Collective Migration"
Metastasis is a complex process that involves the spread of cancer cells from the primary tumor location to distant organs. During metastasis, cancer cells acquire migratory phenotypes, which allow them to detach from the primary tumor and invade the surrounding tissue. Cancer cells migrate through various mechanisms in the tumor microenvironment (TME), including single and collective migration phenotypes. These migration phenotypes are regulated by a complex interplay between the TME, particularly the extracellular matrix (ECM), and the signaling pathways. Here, we developed a computational model using open-source software CompuCell3D (a cellular Potts lattice-based model) that mimics in vitro migration studies of single and collective migration. We consider cancer cells as discrete agents, and their interactions with the TME are simulated in Compucell3D. Using the model, we analyzed the effect of cell-cell adhesion force, non-invasive and invasive phenotypes and structures, and cell-TME interactions in single and collective cell migration. Our aim is to identify key parameters and regulators of cancer metastasis and migration phenotypes. Our model will provide a better understanding of the underlying mechanisms essential for developing more targeted and personalized therapies for cancer metastasis.
ONCO-14
Tyler Simmons
University of Maryland
Poster ID: ONCO-14 (Session: PS01)
"Mathematical Framework of Cellular Exhaustion and the Development of the Tumor-Immune Stalemate"
In response to prolonged tumor-induced stimulation, T cells will dysfunctionally develop into a state of exhaustion. The hypo-functionality of exhausted CD8+ T cells ineffectively combats solid tumors, creating a localized stalemate rather than promoting tumor eradication. In recent years, cellular exhaustion has been a promising target of modern immunotherapy efforts. Exhaustion based therapies attempt to “reverse” this exhaustion, thereby restoring normal effector cell function to better fight the tumor. In this talk we will describe a new mathematical model for modeling the dynamics of exhausted T cells as they interact with cancer. This model follows the development of an exhausted CD8+ T cell population and the subsequent tumor-immune stalemate. Analysis and modeling simulations provide potential future targets for immunotherapy.
ONCO-15
Zeynep Kacar
Univerisity of Maryland
Poster ID: ONCO-15 (Session: PS01)
"Characterization of tumor evolution by functional clonality and phylogenetics in hepatocellular carcinoma"
Hepatocellular carcinoma (HCC) is a molecularly heterogeneous solid malignancy, and its fitness may be shaped by how its tumor cells evolve. However, ability to monitor tumor cell evolution is hampered by the presence of numerous passenger mutations that do not provide any biological consequences. Here, we developed a strategy to determine the tumor clonality of three independent HCC cohorts from 524 patients with diverse etiologies and race/ethnicity by utilizing somatic mutations in cancer driver genes. We identified two main types of tumor evolution, i.e., linear, and non-linear models where non-linear type could be further divide into shallow branching and deep branching. We found that linear evolving HCC is less aggressive than other types. GTF2IRD2B mutations were enriched in HCC with linear evolution while TP53 mutations were the most frequent genetic alterations in HCC with shallow branching and deep branching models. In addition, myeloid cells were more frequently associated with HCC`s non-linear evolution while lymphoid cells were more frequently associated with HCC`s linear evolution. These results suggest that tumor cells and their microenvironment shape the tumor evolution process.
ONCO-16
Javier C. Urcuyo Acevedo
Case Western Reserve University
Poster ID: ONCO-16 (Session: PS01)
"Exploring tumor evolution under the influence of the immune system"
While tumoral heterogeneity play a major role in the development of malignancy, the tumor microenvironment and the initial immune response are equally as important. The immune system is responsible for cancer surveillance and the initial suppression of malignancy. However, with the correct evolutionary mutations, cancer immunoediting can result in immune evasion. Yet, few models incorporate immune components to study the progression and impact of such mutations. In this work, we developed an agent-based model to explore this fitness strategy represented as an evolutionary game. Then, in a phased experimental approach, we plan to validate our model with various immune components, such as CD8+ T cells, NK cells, and other lymphocytes. Ultimately, this work will begin to elucidate the impact of the immune system on cancer evolution and allow us to begin to steer evolution towards more favorable outcomes.
ONCO-17
Gbocho Masato Terasaki
University of California, Merced
Poster ID: ONCO-17 (Session: PS01)
"Merging Traditional Scientific Computing with Data Science to Develop a New Prediction Engine for Brain Cancer"
Glioblastoma multiforme (GBM) is one of the fastest-growing brain tumors and it has very low survival rates. Mathematical modeling can be used to predict the growth and treatment of brain cancer. However, one of the difficulties lies in the ability to estimate patient-specific parameters in the mathematical model from magnetic resonance imaging (MRI) data. We constructed a numerical solver to simulate tumor growth over a realistic 3D brain geometry derived from segmented-MRI. Then, using information about the size of the different glioma sub-regions, we are developing a method that estimates the patient-specific model parameters to inform the forward simulation. Ultimately, we hope to predict the overall survival of a patient from a single pre- operative scan.
ONCO-01
Aaron Li
University of Minnesota
Poster ID: ONCO-01 (Session: PS02)
"A Comparison of Gene Mutation and Amplification-Driven Resistance and Their Impacts on Tumor Recurrence"
Drug sensitive cancer cells often acquire drug resistance, resulting in cancer recurrence despite an initial reduction in tumor size. Two common mechanisms for acquiring drug resistance are point mutation and gene amplification. We propose stochastic multi-type branching process models for each of these mechanisms. Using these models, we derive tumor extinction probabilities and deterministic estimates for the tumor recurrence time, that is, the time when an initially drug sensitive tumor surpasses its original size after developing resistance. For each model, we prove a law of large numbers result regarding the convergence of the stochastic recurrence time to its mean. Additionally, we prove sufficient and necessary conditions for a tumor to escape extinction under the gene amplification model, discuss behavior under biologically relevant parameters, and compare the recurrence time and tumor composition in the mutation and amplification models both analytically and using simulations.
ONCO-02
Alexander Moffett
Northeastern University
Poster ID: ONCO-02 (Session: PS02)
"Modeling the role of immune cell conversion in the tumor-immune microenvironment"
Tumors develop in a complex physical, biochemical, and cellular milieu, referred to as the tumor microenvironment. Of special interest is the set of immune cells that reciprocally interact with the tumor, the tumor-immune microenvironment (TIME). The diversity of cell types and cell-cell interactions in the TIME has led researchers to apply concepts from ecology to describe the dynamics. However, while tumor cells are known to induce immune cells to switch from anti-tumor to pro-tumor phenotypes, this type of ecological interaction has been largely overlooked. To address this gap in cancer modeling, we develop a minimal, ecological model of the TIME with immune cell conversion, to highlight this important interaction and explore its consequences. A key finding is that immune conversion increases the range of parameters supporting a co-existence phase in which the immune system and the tumor reach a stalemate. Our results suggest that further investigation of the consequences of immune cell conversion, using detailed, data-driven models, will be critical for greater understanding of TIME dynamics.
ONCO-03
Austin Hansen
University of California Riverside
Poster ID: ONCO-03 (Session: PS02)
"Computational Modeling of Neural Stem Cell Migration"
Neural stem cells (NSCs) have been shown to be a promising treatment for various brain pathologies due to their ability to migrate directly to the target site, repair damaged tissue, and deliver therapeutic agents. However, the efficacy of such treatments relies on the number and timing of viable cells that are able to reach the injury site. These factors are greatly influenced by different injection strategies as well the complex cytokine dynamics within the brain. For instance, intracranial injections are highly invasive but can be administered next to the site, while intranasal injections can be administered multiple times but require the cells to travel from the olfactory bulb. Furthermore, NSC’s sensitivity to chemoattractants within the brain can alter the path taken and allow for more robust migration. To understand and test the mechanisms that govern NSC migration in a cost effective manner, we build a probabilistic model which accounts for crossing white matter tracts and chemotaxis within the settings of naive rat/mouse brain and TBI. We then use the model to predict the migratory paths in response to different injection strategies/timings as well as explore possible combination therapies to increase NSC arrival rates.
ONCO-04
Brian Johnson
University of California, San Diego
Poster ID: ONCO-04 (Session: PS02)
"Estimating clonal growth rates and the relation to malignancy in human blood"
While evolutionary approaches to medicine hold great potential, measuring evolution is difficult due to experimental constraints and the dynamic nature of biology. It is impossible to continuously observe the evolution of cancer, and obtaining multiple longitudinal samples over time is rare. Advancements in single-cell DNA sequencing have allowed for new evolutionary approaches to studying somatic clonal expansion, which are likely to improve mechanistic understanding of cancer and our ability to effectively prognosticate patients. We present coalescent methods to estimate the growth rate of clones from reconstructed evolutionary trees, eliminating the need for complex simulations. We apply our methods to four recently published single-cell whole genome sequencing datasets, estimating the growth rate of clonal expansions in blood, and validating these estimates with longitudinal data. We show that our estimates lead to new insights on evolutionary parameters, which have implications for early detection of high-risk clones. For example, compared to clones with a single or unknown driver mutation, clones with multiple drivers have increased growth rates (median 0.94 vs. 0.25 per cell per year; p = 1.6 x10^-6). Additionally, patients diagnosed with Myeloproliferative Neoplasm (MPN), a group of malignant conditions characterized by overproduction of blood cells, were found to harbor more aggressively expanding clones (median 0.55 vs. 0.23 per cell per year; p = 0.029) compared to healthy individuals. Further, stratifying patients with MPN by the growth rate of their fittest clone uncovered that higher growth rates are associated with shorter time from clone initiation to MPN diagnosis (median 13.9 vs. 26.4 months; p = 0.0026). As genomic sequencing technology continues to advance, we demonstrate that clonal growth rates can be accurately estimated and have potential for clinical application. To make our methods widely available, we created cloneRate, a user-friendly R package for researchers to apply to their own datasets.
ONCO-05
Clémence Métayer
INSERM U900, Institut Curie, Saint Cloud, France
Poster ID: ONCO-05 (Session: PS02)
"Learning dynamical models of the interactions between the immune receptor NLRP3 and the circadian clock – application to lung cancer"
Lung cancer is a major health problem, with high incidence and mortality rates, due to an absence of effective treatment strategies. In the United States, it is the leading cause of death by cancer, with a 5-year survival of 23% (ACS source). The molecular basis of this disease is complex and heterogeneous, and large inter-patient variability is observed in treatment response, so that it is necessary to consider a mathematical approach to study the processes involved in lung cancer and personalize therapies. In this work, we focus on two deregulated mechanisms in cancer: the immune system and the circadian clock. At the cell level, the circadian clock is a 24-hour biological oscillator that regulates most intracellular processes. It consists of a regulatory network with several intertwine feedback loops that generate sustained oscillations with a period between 20 and 30h. Besides, NLRP3 is a sensor of innate immunity whose role in the immune response has been well studied which was recently identified as an interesting gene altered in lung tumors and predictive of poor prognosis. Previous studies have shown that NLRP3 transcription is regulated indirectly by REV-ERB α (a nuclear receptor of the circadian clock) in macrophages [1][2]. However, the links between NLRP3 and the circadian clock and in particular the impact of the clock on the function of NLRP3 have been very rarely investigated. Our goal is then to characterize the interactions between NLRP3 and the circadian clock that are emerging as major components in the pathophysiology of lung cancer. To this end, we have undertaken a combined experimental and mathematical approach. We have studied the interactions of NLRP3 and the circadian clock in human bronchial epithelial cells (HBEC) which were synchronized by serum shock. Transcriptomics (RNA-Sequencing) and proteomics (Mass Spectrometry) data as well as intracellular localization (nucleus/cytoplasm) were assessed. Clock gene components were defined using the Reactome database (v84). Circadian rhythms were studied using cosine wave fitting and using CMAES for the minimization task. Model learning method was developed to automatically learn the structure of quantitative systems biology models based on ordinary differential equations from multimodal data. Parameter estimation was performed using a modified least-square approach using CMAES for minimization. The analysis of mRNA levels of 67 clock genes revealed a functional clock in HBEC cells with a period of 30h+/-2h . NLRP3 can interact with clock proteins and the data suggest that they could regulate the intracellular localization of NLRP3 to orchestrate its functions. On the other hand, loss of NLRP3 expression may disrupt the circadian regulation necessary for normal lung function. An existing circadian clock model [3] using ordinary differential equations (ODE) was extended by adding equations describing the influence of the clock on NLRP3 transcription and interactions of clock and NLRP3 proteins. As a start, a collection of models were considered that included a single additional reaction as compared to the initial clock model. Datasets used for the fit were: mRNA levels of 7 clock genes, protein level of 7 clock genes and circadian rhythms of nucleus/cytoplasm localization of NLRP3, BMAL1, PER2 and CRY1. A systematic fit of each model was performed which allowed to eliminate unlikely reactions. Models involving more than one additional reaction are being investigated. Such model learning pipeline will help prioritize future experiments to fully determine NLRP3 interactions with the clock and identify potential drug targets to restore NLRP3 functions in NLRP3-altered cancer cells.
[1] Pourcet, B., Zecchin, M., Ferri, L., Beauchamp, J., Sitaula, S., Billon, C., ... & Duez, H. M. (2018). Nuclear receptor subfamily 1 group D member 1 regulates circadian activity of NLRP3 inflammasome to reduce the severity of fulminant hepatitis in mice. Gastroenterology, 154(5), 1449-1464. [2] Wang, S., Lin, Y., Yuan, X., Li, F., Guo, L., & Wu, B. (2018). REV-ERBα integrates colon clock with experimental colitis through regulation of NF-κB/NLRP3 axis. Nature communications, 9(1), 1-12. [3] J. Hesse, J. Martinelli, O. Aboumanify, A. Ballesta, and A. Relogio. A mathematical model of the circadian clock and drug pharmacology to optimize irinotecan administration timing in colorectal cancer. Computational and structural biotechnology journal, 19:5170–5183, 2021.
ONCO-06
Jonathan Rodriguez
University of California, Irvine
Poster ID: ONCO-06 (Session: PS02)
"Predictive nonlinear modeling of malignant myelopoiesis and tyrosine kinase inhibitor therapy"
Chronic myeloid leukemia (CML) is a blood cancer characterized by dysregulated production of maturing myeloid cells driven by the product of the Philadelphia chromosome, the BCR-ABL1 tyrosine kinase. Tyrosine kinase inhibitors (TKI) have proved effective in treating CML but there is still a cohort of patients who do not respond to TKI therapy even in the absence of mutations in the BCR-ABL1 kinase domain that mediate drug resistance. To discover novel strategies to improve TKI therapy in CML, we developed a nonlinear mathematical model of CML hematopoiesis that incorporates feedback control and lineage branching. Cell-cell interactions were constrained using an automated model selection method together with previous observations and new in vivo data from a chimeric BCR-ABL1 transgenic mouse model of CML. The resulting quantitative model captures the dynamics of normal and CML cells at various stages of the disease, exhibits variable responses to TKI treatment, predicts key factors of refractory response to TKI treatment, and predicts potential combination therapy efficacy. Recent experiments reveal that interactions and competition between different cellular compartments and between normal and BCR-ABL1-expressing cells form a threshold that determines whether the malignant cells can expand and cause leukemia. To capture these experimental dynamics, we found it necessary to incorporate additional biological factors through the introduction of new cell types and interactions. We applied an adapted model selection scheme to explore the unknown cell-cell interaction space and find subsets of models consistent with experimental dynamics. We analyzed common motifs across experimentally consistent models and identified interactions as targets for experimental design to further narrow the valid models.
ONCO-07
Kailei Liu
University at Buffalo, The State University of New York, Buffalo, NY
Poster ID: ONCO-07 (Session: PS02)
"Computational modeling of cell migration in complex chemokine environments"
In recent decades, research on the active expression and regulatory effects of chemokines in cancer and immune cells has made the chemokine system an emerging target of immunotherapy. Alteration in chemokine environments is expected during immunotherapy, emphasizing the importance of understanding cell migration in complex chemokine environments. The complex signaling network formed by chemokines and cognate receptors regulates diverse tumor and immune cell activities, including leukocyte recruitment, angiogenesis, tumor growth, proliferation, and metastasis. We built 2D & 3D agent-based models with Compucell3D (a cellular Potts lattice-based model) to simulate the physiological response, especially cell migration, of tumor and immune cells towards complex chemokine settings. The 2D model is used to understand the mechanisms of cell chemotaxis, monomer-dimer equilibrium of certain chemokines, and competition between different pairs of chemokines and cognate receptors. The 3D model simulates and predicts an in vitro transwell experiment where cells have more realistic biomechanics of neighboring cells and tissue-mimic biomaterials. Using the models, we investigated how chemokine concentration, chemotactic force, environment composition, energy term that governs random walk, and membrane properties can influence cell migration. Results from this study will be used to build new agent-based models to simulate in vivo cancer pathology and therapy, considering cells, chemokines, and tissue microenvironments.
ONCO-08
Khaphetsi Joseph Mahasa
National University of Lesotho
Poster ID: ONCO-08 (Session: PS02)
"CD8+ T cells against circulating tumor cells coated with platelets: Insights from a mathematical model"
Cancer metastasis accounts for many cancer-related deaths worldwide. Metastasis occurs when the primary tumor sheds cells into the blood and/ or lymphatic circulation, thereby becoming circulating tumor cells (CTCs) that transverse through the circulatory system, extravasate the circulation and establish a secondary distant tumor. CTCs transition through the blood system, which has a plethora of supportive and antitumoral immune cells, represents one of the major metastatic hurdles that are not yet fully deciphered. Upon their entry into the blood stream, CTCs interact with platelets which shield them from the recognition by immune cells, including natural killer cells and CD8+ T cells. Platelet binding to CTCs also enhances CTC arrest in the vascular endothelial walls and subsequent extravasation. On the other hand, activated circulating CD8+ T cells, are able to recognise and attack the arrested CTCs prior to their extravasation. Thus, understanding how the dynamic interactions between CTCs, platelets and CD8+ T cells eventually result in secondary metastatic tumor emergence is a key challenge. Here, through a simple mathematical model of ordinary differential equations, we provide our perspective on how CTCs mechanistically evade the CD8+ T cell cytotoxicity. To achieve this, we aim to (a) describe how the intrinsic growth of the primary tumor, and subsequent dissimination of CTCs, link to the secondary establishment of distant tumor cell population; (b) illustrate how the intravascular proliferation of arrested CTCs within the circulation contributes to the possibility of secondary metastasis; (c) describe possible mechanisms underlying the antitumoral activity of CD8+T cells in inhibiting metastatic potential of CTCs; (d) discuss how simple treatment scenarios can be employed to minimize a further spread of CTCs within the circulation, by focusing on the CTCs disseminated from the primary tumor, rather than the secondary metastatic tumor. Last, we also provide a comprehensive mathematical stability analyses to assess different treatment scenarios that can hamper CTCs survival and highlight the significant role of mathematical modeling in clinical oncology.
ONCO-09
Matthew Froid
H. Lee Moffitt Cancer Center and Research Institute
Poster ID: ONCO-09 (Session: PS02)
"A Hybrid Modeling Approach Illuminates Physical and Genetic Factors Contributing to Resistance in the AML Bone Marrow Niche"
BACKGROUND: Acute myeloid leukemia (AML) outcomes remain poor, likely due to treatment-resistant leukemic stem cells (LSCs). Evidence suggests resistance to tyrosine kinase inhibitors (TKIs) depends on the bone marrow’s vascular plasticity via the Janus Phenomena. The opposing “faces” of the Janus Phenomena are the initial beneficial cytoreduction of blast cells, followed by revascularization of the BM by endothelial cells, the expansion of LSCs, and then relapse. This is partially mediated through miR-126 over-expression upon which the endothelial cells are dependent to revascularize the BM and to shelter LSCs. To mitigate the Janus Phenomena, miRisten, a miR-126 expression inhibitor, was developed. To understand the interactions of TKI with miRisten, we used experimental data to inform an agent-based model (ABM) recapitulating the Janus Phenomena to explore how different vasculature architectures and drug scheduling can prevent relapse. In addition to the BM structure, we explored common interactors among genes linked to both drugs (AC220, a TKI, and miRisten). METHODS: Using an on-lattice 2D ABM containing three agents (EC cells, LSCs, and blast cells), we modeled 16 different vascular architectures informed from mouse tibias post-TKI treatment. We tested three conditions: TKI, TKI + miRisten, and TKI + miRisten pre-treatment. K-means clustering and PCA were performed to determine relationships among the varying vascular architectures. A protein-protein interaction (PPI) network based on publicly available data for the proteins affected by both drugs was constructed. We used several centrality measurements to determine the nodes in the directed graph that have the largest role in the connectivity of the network. Next we constructed a graph neural network to classify proteins linked to either the targets of AC220 or miRisten with limited or tenuous experimental validation. RESULTS: The optimal dose strategy to prevent relapse was two-weeks of pre-treatment with mRristen before TKI administration. Additionally, vascular architectures spanning the entire domain of the ABM consistently prevented relapse during treatment. Interestingly within the drugs’ PPI, miR-206 (a known tumor suppressor linked to angiogenesis and an indirect regulator of miR-126) was the node with the highest degree centrality. CONCLUSION: To prevent AML relapse under TKI, miRisten should be given for two weeks before starting TKI. Additionally, both the physical structure of the vasculature and the protein-protein interactors contribute to resistance.
ONCO-10
Megan LaMonica
The University of Texas at Austin
Poster ID: ONCO-10 (Session: PS02)
"Investigating limits of predictability of a 3D reaction-diffusion glioblastoma model"
Introduction: Predictive mathematical models of glioma growth and therapy response can be informed with quantitative magnetic resonance imaging (MRI) data [1]. However, it is unclear what quantity and quality of longitudinal MRI data are required for accurate model calibration and prediction. To address this uncertainty, we utilize a novel in silico framework that explores the predictability limits of a spatiotemporal reaction-diffusion glioma model by quantifying how different combinations of signal-to-noise ratio (SNR), spatial resolution (SR), and temporal resolution (TR) in initial murine MRI data affect accuracy of parameter calibration and tumor growth prediction. Methods: We have developed a two-species reaction-diffusion glioma model that describes the spatiotemporal evolution of tumor cellularity and vascularity [2]. The initial cellularity and vascularity conditions that inform the model are estimated from quantitative MRI data. Different combinations of SNR, SR, and TR are applied to the initial conditions. TR is varied by changing the quantity and spacing of the MRI data used to inform parameter calibration. We apply the model to a spatially heterogeneous rat tumor in a simulated brain tissue domain [3]. We then solve the model via the finite difference method and calibrate model parameters using the Levenberg-Marquardt algorithm (N = 50 in silico replicates). We report the mean and standard deviation of each model parameter error as well as error in longitudinal tumor volume prediction for each tested combination of SNR, SR, and TR. Results and future directions: Low SR (voxel volume 0.500 mm3), experimentally relevant SR (voxel volume 0.063 mm3), and high SR (voxel volume 0.008 mm3) conditions are evaluated across a range of TR and SNR for all model parameters. The worst TR case uses two calibration timepoints, 96 hours apart; the experimentally relevant case uses three timepoints, 48 hours apart; and finally, the best case uses five timepoints, 24 hours apart. SNR is tested from 5 to 160. At an experimentally relevant SNR of 40, calibrated parameter percent error (PE) in tumor diffusion and proliferation falls by approximately 75% as SR improves. PE falls by approximately 37% as TR improves in this same SNR case. With SR held constant at the 0.063 mm3 voxel volume SR condition, PE decreases by approximately 40% between an SNR of 20 and 40, with little improvement seen past an SNR of 80. Global concordance correlation coefficients and Dice similarity coefficients were relatively consistent across all tested combinations. The next step will be to evaluate the model with different ground truth tumors in order to recommend target combinations of SR, TR, and SNR for a wider range of tumor types and experimental conditions. Funding: CPRIT RR160005, RP220225; NIH R01CA235800, U24CA226110, U01CA174706. References: [1] Hormuth et al., Advanced Drug Delivery Reviews, 187(114367), 2022. [2] Hormuth et al., Cancers, 13(8), 2021. [3] Hormuth et al., Ann. Biomed. Eng., 47(7), 2019.
ONCO-11
Nadia Wright
Arizona State University
Poster ID: ONCO-11 (Session: PS02)
"Castration resistance in prostate cancer arises through both natural selection and phenotypic plasticity"
Recurrent prostate tumors are commonly treated with a total androgen blockade via chemical castration. In turn, cancerous cells have been known to respond with an evolutionary up-regulation of androgen receptors (AR), thus prolonging cell proliferation and delaying apoptosis. Prostate epithelial cancers treated with androgen ablation therapy invariably become castration resistant. However, the primary mechanism remains unknown. Suggested hypotheses include phenotypic plasticity and natural selection. Here we show that castration resistance in prostate cancers treated with androgen ablation arises through natural selection acting on phenotypic plasticity. We found that tumor aggressiveness, measured as growth rate of serum concentration of prostate specific antigen (PSA), correlates positively with the number of treatment cycles. Additionally, we found a signal of increasing tumor aggressiveness with cycle in both on and off-treatment phases. This result argues against the plasticity hypothesis and is consistent with evolution by natural selection. If plasticity were the mechanism, then tumor aggressiveness would not correlate with cycle. This result can help inform clinical management of prostate cancer treated with androgen ablation. Identification of the exact evolutionary mechanism will almost certainly yield insight into more efficacious treatment schedules, and drug combinations, while maintaining patient quality of life and delaying the onset of castration resistance.
ONCO-12
Rafael R Bravo
Moffitt Cancer Center
Poster ID: ONCO-12 (Session: PS02)
"Using MRI scans to predict tumor margin propagation in GBM under immunotherapy"
An active area of GBM research is identifying how properties of the brain tissue around the tumor as detected by MRI impact tumor growth and treatment response. Such information could be useful in determining surgical margins and optimizing patient specific treatment selection.
To answer this question, we developed a tumor margin propagating algorithm in which the margin growth or shrinking rate can be locally accelerated or slowed according to T1, T1-post, T2, ADC, and FLAIR scan values. We measured how well the growing/shrinking tumor margin starting from a patient scan overlaps with the tumor margin from the subsequent patient scan with these local rate adjustments.
We applied this approach to MRI sequences from 32 patients treated with hypofractionated stereotactic radiotherapy, bevacizumab and pembrolizumab at Moffitt Cancer Center. We found that the tumors tend have affinity for high-FLAIR regions in most cases, and that tumors that are growing tend to have affinity for high-T1 regions, and tumors that are shrinking tend to avoid high-T1 regions. Once our findings from this project are fully developed, they may assist future modeling efforts to predict tumor proliferation and response to immunotherapy in GBM.
ONCO-13
Stefano Pasetto
Moffitt Cancer Center
Poster ID: ONCO-13 (Session: PS02)
"Calibrating tumor growth and invasion parameters with spectral-spatial Analysis of cancer biopsy tissues"
Predictive modeling in oncology is a growing field. The calibration of mathematical model parameters based on limited clinical data is critical to reliable predictions per-patient basis. One omnipresent mathematical model is the reaction-diffusion equation, which has been shown to simulate and predict clinical parameters in different cancer types. Here, we focus on analyzing cell-level data routinely obtained from tissue biopsies at diagnosis for most cancers. We analyze the spatial architecture in biopsy tissues stained with multiplex immunofluorescence. We derive the two-point correlation function and the corresponding spatial power spectral distribution. We show that the data-deduced spatial power spectral distribution can fit the spatial power spectrum of the solution of the reaction-diffusion equation, thereby identifying patient-specific tumor growth and invasion rates from a single, routinely collected clinical tissue. This novel approach is essential for model-parameter-inference for tumor infiltration, which may ultimately be used to inform clinical treatments.
ONCO-14
Megan LaMonica
The University of Texas at Austin
Poster ID: ONCO-14 (Session: PS02)
"Investigating the impact tumor heterogeneity has on patient response to radiotherapy via mathematical modeling"
The overall purpose of this study is to determine how different assumptions of radiotherapy efficacy affect predictions of tumor cell count using a biology-based mathematical model describing the spatiotemporal evolution of tumor growth and response to radiotherapy. Models seeking to predict patient-specific response have yet to characterize intratumoral heterogeneity in response to radiotherapy. To address this limitation, we acquired quantitative magnetic resonance imaging (MRI) data on four patients with high-grade gliomas at the MD Anderson Cancer Center being treated with fractionated radiotherapy. This longitudinal data was then used to inform a two-species mechanically-coupled reaction diffusion model [1] describing the spatiotemporal change of tumor growth and response to therapy. Tumor cell proliferation rates, tumor diffusion coefficients, and response to radiotherapy (estimated as the surviving fraction following a single radiotherapy session) were calibrated from data up to 1-month post-radiotherapy using the Levenberg-Marquardt approach in MATLAB. With these patient-specific calibrated parameters, our model simulated tumor growth and response assuming treatment efficacy varies homogeneously (globally) or heterogeneously (as a function of vasculature and cell density). We calculated and compared the percent change in tumor cell count three months after initial treatment for surviving fractions of 0.2 to 1 (in increments of 0.05) for four patients. Treatment response as observed at 3-months post-radiotherapy varied greatly (from eradication to residual disease) depending on each assumption on the spatial variations in efficacy. For example, a surviving fraction of 0.6 resulted in complete eradication of the tumor under both homogenous and heterogenous (cell density) assumptions. However, when radiotherapy efficacy was related to vasculature, only an average 55% decrease in tumor cell count was observed. Thus, we have developed an approach to quantify the impact of different assumptions of heterogeneity in response to radiation on percent change in tumor cell count. Future efforts will extend this approach to a larger cohort of patients.
ONCO-15
Yixuan Wang
University of Michigan
Poster ID: ONCO-15 (Session: PS02)
"Modeling CTL-mediated Tumor Cell Death Mechanisms and the Activity of Immune Checkpoints in Immunotherapy"
Immunotherapy has dramatically transformed the cancer treatment landscape. Of the variety of types of immunotherapies available, immune checkpoint inhibitors (ICIs) have gained the spotlight. Although ICIs have shown promising results for some patients, the low response rates in many cancers highlight the challenges of using immune checkpoint blockade as an effective treatment. Cytotoxic T lymphocytes (CTLs) execute their cell-killing function via two distinct mechanisms. The first process is fast-acting and perforin/granzyme-mediated, and the second is a slower, Fas ligand (FasL)-driven killing mechanism. There is also evidence suggesting that the preferred killing mechanism by CTLs depends on the antigenicity of tumor cells. To determine the key factors affecting responses to checkpoint blockade therapy, we constructed an ordinary differential equation model describing in vivo tumor-immune dynamics in the presence of active or blocked PD-1/PD- L1 immune checkpoint. Specifically, we analyzed which aspects of the tumor-immune landscape affect the response to ICIs with endpoints of tumor size and composition in the short and long term. By generating a virtual cohort with heterogeneous tumor and immune attributes, we also simulated the therapeutic outcomes of immune checkpoint blockade in a largely diverse population. In this way, we identified key tumor and immune characteristics that are associated with tumor elimination, dormancy and escape. Our analysis sheds light on which fraction of a population potentially responds well to ICIs and ways to enhance therapeutic outcomes with combination therapy.
ONCO-16
Megan LaMonica
The University of Texas at Austin
Poster ID: ONCO-16 (Session: PS02)
"Investigating limits of predictability of a 3D reaction-diffusion glioblastoma model"
Introduction: Predictive mathematical models of glioma growth and therapy response can be informed with quantitative magnetic resonance imaging (MRI) data [1]. However, it is unclear what quantity and quality of longitudinal MRI data are required for accurate model calibration and prediction. To address this uncertainty, we utilize a novel in silico framework that explores the predictability limits of a spatiotemporal reaction-diffusion glioma model by quantifying how different combinations of signal-to-noise ratio (SNR), spatial resolution (SR), and temporal resolution (TR) in initial murine MRI data affect accuracy of parameter calibration and tumor growth prediction. Methods: We have developed a two-species reaction-diffusion glioma model that describes the spatiotemporal evolution of tumor cellularity and vascularity [2]. The initial cellularity and vascularity conditions that inform the model are estimated from quantitative MRI data. Different combinations of SNR, SR, and TR are applied to the initial conditions. TR is varied by changing the quantity and spacing of the MRI data used to inform parameter calibration. We apply the model to a spatially heterogeneous rat tumor in a simulated brain tissue domain [3]. We then solve the model via the finite difference method and calibrate model parameters using the Levenberg-Marquardt algorithm (N = 50 in silico replicates). We report the mean and standard deviation of each model parameter error as well as error in longitudinal tumor volume prediction for each tested combination of SNR, SR, and TR. Results and future directions: Low SR (voxel volume 0.500 mm3), experimentally relevant SR (voxel volume 0.063 mm3), and high SR (voxel volume 0.008 mm3) conditions are evaluated across a range of TR and SNR for all model parameters. The worst TR case uses two calibration timepoints, 96 hours apart; the experimentally relevant case uses three timepoints, 48 hours apart; and finally, the best case uses five timepoints, 24 hours apart. SNR is tested from 5 to 160. At an experimentally relevant SNR of 40, calibrated parameter percent error (PE) in tumor diffusion and proliferation falls by approximately 75% as SR improves. PE falls by approximately 37% as TR improves in this same SNR case. With SR held constant at the 0.063 mm3 voxel volume SR condition, PE decreases by approximately 40% between an SNR of 20 and 40, with little improvement seen past an SNR of 80. Global concordance correlation coefficients and Dice similarity coefficients were relatively consistent across all tested combinations. The next step will be to evaluate the model with different ground truth tumors in order to recommend target combinations of SR, TR, and SNR for a wider range of tumor types and experimental conditions. Funding: CPRIT RR160005, RP220225; NIH R01CA235800, U24CA226110, U01CA174706. References: [1] Hormuth et al., Advanced Drug Delivery Reviews, 187(114367), 2022. [2] Hormuth et al., Cancers, 13(8), 2021. [3] Hormuth et al., Ann. Biomed. Eng., 47(7), 2019.
ONCO-17
Gustav Lindwall
Chalmers University of Technology, Gothenburg, Sweden
Poster ID: ONCO-17 (Session: PS02)
"Statistical inference on interacting particle systems with applications to cancer biology "
In this poster, I will summarize the content of my PhD thesis. The main concern of my studies has been mathematical modelling of in vitro cancer cell migration. Along with themodelling, an array of Bayesian statistical inference algorithms for key parameters in the models are presented. The guiding principle behind my research interest is that solid models derived from physical principles can aid in the understanding of how cancer cells interact with one another. The subsequent clinical applications of this research can for example be profiling of cells sampled from a specific patient, aiding the physician in choice of clinical intervention. My model paradigm of choice are agent-based models, where every single cell in the sample is given consideration as an agent. The fundamental building block is a set of stochasticdifferential equations (SDE:s) describing the current location of all cells. We also incorporate cell proliferation into our model, every cell divides or die according to a non-homogeneous Poisson process depending the state of the population.