Models from Experiments: Combinatorial Drug Perturbations of Cancer Cells
Sven Nelander,Weiqing Wang,Bengt‐Olof Nilsson,Qing‐Bai She,Christine A. Pratilas,Neal Rosen,Peter Gennemark,Chris Sander
DOI: https://doi.org/10.1038/msb.2008.53
IF: 13.068
2008-01-01
Molecular Systems Biology
Abstract:Report2 September 2008Open Access Models from experiments: combinatorial drug perturbations of cancer cells Sven Nelander Corresponding Author Sven Nelander Computational Biology center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Weiqing Wang Weiqing Wang Computational Biology center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Björn Nilsson Björn Nilsson Department of Clinical Genetics, Lund University Hospital, Lund, Sweden Search for more papers by this author Qing-Bai She Qing-Bai She Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Christine Pratilas Christine Pratilas Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Neal Rosen Neal Rosen Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Peter Gennemark Peter Gennemark Department of Mathematical Sciences, University of Gothenburg, Gothenburg, Sweden Search for more papers by this author Chris Sander Chris Sander Computational Biology center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Sven Nelander Corresponding Author Sven Nelander Computational Biology center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Weiqing Wang Weiqing Wang Computational Biology center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Björn Nilsson Björn Nilsson Department of Clinical Genetics, Lund University Hospital, Lund, Sweden Search for more papers by this author Qing-Bai She Qing-Bai She Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Christine Pratilas Christine Pratilas Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Neal Rosen Neal Rosen Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Peter Gennemark Peter Gennemark Department of Mathematical Sciences, University of Gothenburg, Gothenburg, Sweden Search for more papers by this author Chris Sander Chris Sander Computational Biology center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA Search for more papers by this author Author Information Sven Nelander 1, Weiqing Wang1, Björn Nilsson2, Qing-Bai She3, Christine Pratilas3, Neal Rosen3, Peter Gennemark4 and Chris Sander1 1Computational Biology center, Memorial Sloan-Kettering Cancer Center, New York, NY, USA 2Department of Clinical Genetics, Lund University Hospital, Lund, Sweden 3Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA 4Department of Mathematical Sciences, University of Gothenburg, Gothenburg, Sweden *Corresponding author. E-mail correspondence reaches the principal authors at multiple-perturbation>at0 means ‘node j activates node i’, whereas wij<0 corresponds to inhibition.) Furthermore, αi>0 represents the tendency of the system to return to the initial state (yi=0); βi>0 are constants and ϕi is a transfer function capable of capturing both switch-like behavior and bounded reaction rates. Examples of such functions include sigmoid functions, piece-wise linear approximations of sigmoids or biochemically motivated approximations such as the Hill or Michaelis–Menten equations (Materials and methods). Application of nonlinear MIMO models to combinatorial perturbation experiments We developed computer algorithms to infer nonlinear models of the above type from experimental data, as specified by the best-performing values of the coupling parameters wij and other parameters. As detailed in Materials and methods, the current implementation of our approach consists of the following steps. First, the system of interest is subjected to a set of independent single or multiple target perturbation experiments; and, for each perturbation vector (time-independent instance of u), a readout vector (steady-state instance of y) is recorded. Second, we infer a nonlinear model that best reproduces the experimental data (Materials and methods). Specifically, we rely on parameter estimation techniques for feedback systems to find a model that minimizes a quadratic error term between observed and predicted readouts, subject to simplicity constraints on the number of interactions in the system. Third, the fitted model can be used to predict the system's response to unseen perturbations (for example, combinations of drugs), and to gain new insight into the system's architecture. Testing modeling power for combinatorial perturbations in breast cancer cells Dual drug perturbation experiments in MCF7 breast cancer cells To directly test the power of the approach, we performed an independent experimental study in MCF7 human breast carcinoma cells. As perturbants of the system, we chose compounds targeting EGFR (ZD1839), mTOR (rapamycin), MEK (PD0325901), PKC-δ (rottlerin), PI3 kinase (LY294002) and IGF1R (A12 anti-IGF1R inhibitory antibody). As relevant readouts of molecular and phenotypic responses, we chose phospho-protein levels of seven regulators of survival, proliferation and protein synthesis (p-AKT-S473, p-ERK-T202/Y204, p-MEK-S217/S221, p-eIF4E-S209, p-c-RAF-S289/S296/S301, p-P70S6K-S371 and pS6-S235/S236) as well as flow cytometric observation of two phenotypic processes (cell cycle arrest and apoptosis) (Figure 2). Inhibitors were administered singly and in pairs, followed by EGF stimulation. When recording responses of protein phosphorylation, we used the average response at 5 and 30 min as the surrogate for steady-state values. To build models, we represented the state of each of the above perturbation targets (signaling proteins), as well as each of the readouts, by one state variable yi. We then used the proposed optimization procedure (Materials and methods) to estimate the coupling parameters wij and other parameters, resulting in predictive models of response in terms of these system variables. Figure 2.Breast cancer cells as a multiple input–multiple output system. To generate data for model construction, we treated human MCF7 breast tumor cell lines with one natural ligand (epidermal growth factor (EGF)) and six inhibitors, singly and in combination. The treatment protocol used EGF, an IGF1 receptor inhibitory antibody (A12) and inhibitors of the signaling molecules EGFR, PI3K, mTOR, PKC-δ and MEK. The inhibitors are ZD1839, LY294002, rapamycin, rottlerin and PD0325901. In the perturbation matrix (top panel, columns=experiments, rows=perturbations), a blue box indicates the presence of a particular perturbation and white indicates absence. For each treatment, we used western blots to detect levels of the proteins phospho-AKT, phospho-ERK, phospho-MEK, phospho-eIF4E, phospho-c-RAF, phospho-p70S6K and phospho pS6. We used a FACS-based assay to quantify apoptosis (measured as the ‘sub-G1 fraction,’ Materials and methods) and G1 arrest (measured as the G1 fraction). Here, representative flow histograms depicting cell cycle distribution in MCF7 cultures treated with or without drug are shown (one experiment is shown, see Supplementary information for all measurements). Evaluation of predictive power: After model construction based on these experiments, we see good agreement between experimental observation of the response of the MCF7 cell line to the 21 different perturbations (top, columns=experiments, rows=readouts) and the model prediction (bottom). Statistical assessment is in Supplementary Table 1. For each readout, we quantify the system's response by the phenotypic index defined as log relative response in treated versus untreated cells. For some drug combinations, the phenotypic readout increases as a result of perturbation (orange), for others it decreases (blue). Download figure Download PowerPoint Quantitative prediction of system response We first assessed the predictive power of the derived models using leave-one-out cross-validation, in which one pair perturbation is left out of the analysis and then its effect predicted from information gained from all other perturbations. The resulting predictions were reasonably accurate for the nine different readouts. The best prediction was obtained for p-S6 phospho-protein levels (cross-validation error CV=0.02, Pearson correlation r=0.96) and the weakest for the G1 arrest phenotype (CV=0.07, r=0.45) (Figure 2 and Supplementary Table 1). We directly compared the performance of our modeling approach to one using a corresponding set of linear differential equations with the same optimization procedure. By comparison, predictions using the nonlinear approach agreed better with experimental observations for eight of the nine readouts. Using the nonlinear modeling approach, the prediction error was lower by up to 50% with correspondingly better correlation between predictions and experimental observations (Supplementary Table 1). Thus, we conclude that our method is capable of deriving reasonably accurate network models for the input–output behavior of MCF7 cells with respect to the readouts used. Detection of key regulatory mechanisms without prior knowledge From a set of perturbation experiments, how can one deduce the logical network structure of activating and inhibiting interactions between the key molecular components, similar to the familiar pathway diagrams in publications summarizing a set of molecular biological experiments? Here, we use the derived network models with the smallest global error (Etotal=ESSQ+λESTRUCT, Materials and methods) to infer causal connectivity diagrams. The inference is based on the assumption that interactions in sufficiently simple models that fit experimental observations, called ‘good’ models, represent an underlying causal relationship between system components modeled by the system variables yi. Such a relationship can be either an indirect regulatory effect or a direct physical interaction that would be observable in vitro with purified components. Using our Monte Carlo algorithm, we generated a population of 450 good models from the MCF7 dual drug perturbation experiments. From these, we assessed the statistical significance of the individual interactions both in terms of a posterior probability (which is obtained directly from the Monte Carlo process, see Materials and methods) and a 90% confidence interval constructed by boot-strapping simulations (Table I). We now discuss the connectivity of the best model, i.e. the one with the smallest error (schema in Figure 3, explicit equations in Materials and methods) relative to the known biology of regulatory pathways in the MCF7 breast cancer cell line. Figure 3.Use of MIMO models to infer regulatory interactions in breast cancer cells. The interaction matrix wij from a set of good models can be used to infer regulatory interactions (squares=inputs; circles=internal system variables and other observables). Positive wij means activation and negative wij means inhibition of the target. Interestingly, some of the interactions are well known in MCF7 cells (green arcs) and others constitute predictions (orange arcs). See Table I for functional comments on interactions. No underlying pathway model was used—the network is a straightforward interpretation of the optimized model parameters wij. The EGFR → MEK → ERK and PI3K → AKT, canonical pathways are identified. Also, note the detection of self-inhibitory interactions in MEK/ERK signaling, identification of eIF4E and AKT as direct regulators of apoptosis and G1 arrest. Download figure Download PowerPoint Table 1. Statistical assessment of inferred interactions in MCF7 cells Regulator Target Mean interaction 90% CI Posterior probability (%) Comment EGFR → MEK 0.78 (0.67, 0.88) 99 0 Activation of MEK by RTK signaling MEK → p-ERK 0.39 (0.26, 0.48) 99 0 MAPK signaling PI3K → p-AKT 0.57 (0.47, 0.69) 93 0 PI3K-dependent activation of AKT PKC-δ → p-RAF 0.25 (0.17, 0.32) 71 0 Prediction mTOR → p-P70S6K 0.25 (0.20, 0.29) 41 0 mTOR signaling pathway p-ERK → p-eIF4E 0.19 (0.14, 0.25) 38 0 Prediction p-ERK → p-ERK 0.29 (0.22, 0.40) 35 0 Prediction p-P70S6K → p-S6 0.54 (0.53, 0.56) 20 0 S6 kinase activates S6 Apoptosis → G1 arrest 0.37 (0.30, 0.44) 20 0 Prediction G1 arrest → EGFR 0.39 (0.29, 0.49) 13 0 Low significance prediction MEK → p-RAF 0.29 (0.16, 0.41) 13 0 Low significance prediction p-eIF4E → PI3K 0.05 (0.02, 0.08) 13 1 Low significance prediction p-AKT Apoptosis −0.39 (−0.43, −0.36) 0 77 AKT controls survival in MCF7 cells PKC-δ G1 arrest −0.12 (−0.20, −0.06) 0 41 Prediction p-ERK EGFR −0.28 (−0.42, −0.17) 0 36 MAPK negative feedback loop G1 arrest p-eIF4E −0.33 (−0.36, −0.30) 0 32 Predicted bi-stable regulation p-eIF4E G1 arrest −0.19 (−0.25, −0.12) 0 20 Predicted bi-stable regulation Apoptosis p-P70S6K −0.39 (−0.42, −0.36) 0 19 Low significance prediction IGF1R G1 arrest −0.06 (−0.14, −0.01) 0 16 Low significance prediction G1 arrest p-ERK −0.09 (−0.16, −0.01) 1 15 Low significance prediction mTOR Apoptosis −0.07 (−0.12, −0.02) 1 12 Low significance prediction MEK MEK −0.13 (−0.21, −0.07) 0 8 Low significance prediction EGFR p-RAF −0.03 (−0.11, −0.04) 6 2 Low significance prediction Statistical assessment of inferred molecular interactions as shown in Figure 3. Column ‘Mean Wij’ shows the average interaction strength between the target and the regulator, as obtained from 200 bootstrapping simulations (see Supplementary information). 90% confidence intervals (CI) for the interaction strength are shown. The left column of posterior probabilities shows the fraction of models with a positive regulation in the Monte Carlo simulation. The right column shows the fraction of models with negative regulation (for instance, inhibition of apoptosis by p-AKT was present in 77% of models). The names p-P70S6K and p-p70S6K are synonymous. Interpretation of derived network structure In comparing the inferred connectivity with mechanisms known to occur in MCF7 cells (Table I), two caveats are important. (1) The logical nodes in our models are defined precisely as the perturbed and observed molecular species, i.e. the targets of drug perturbation and the targets of specific observed antibody reactions, and may not be exactly identical to a single molecular species. For example, ‘EGFR’ refers to the direct target(s) of activation by EGF and of inhibition by the drug ZD1839, and these two are assumed to be identical. (2) The models make no reference to unperturbed or unobserved nodes, e.g. whereas p-AKT is in the network model, the unphosphorylated AKT is not. With these caveats in mind, one can use the models both for confirmation and prediction of interactions. Of the 23 interactions in the best model, 14 had a posterior probability in the range of 20–99% (Table I). Of these, several statistically robust interactions clearly confirm canonical pathway structures. (i) The MAPK cascade downstream of the EGF receptor is detected as a chain of interactions between EGFR, MEK and ERK (Figure 3 and Table I). (ii) The negative feedback regulation of MAPK signaling is captured as negative interaction from ERK to EGFR, and as a moderately significant self-inhibition of MEK (see Discussion). (iii) PI3K-dependent signaling and the tendency for MCF7 cells to be dependent on AKT activation for survival are detected as interactions between PI3K, AKT and the apoptosis phenotype. (iv) The model inference that apoptosis is controlled by p-AKT, but not p-ERK, is in agreement with previous results in MCF7 cells (Simstein et al, 2003; DeFeo-Jones et al, 2005). (v) mTOR downstream signaling is detected as interactions between mTOR, p70S6K and ribosomal S6 protein (Mingo-Sion et al, 2005). The derivation of these expected interactions from a small set of perturbation experiments, without prior pathway knowledge, underscores the non-trivial value of the model building approach and provides some confidence in the concrete predictions of logical regulatory interactions for MCF7 cells (Table I), which are discussed below. Discussion In summary, our evaluation in breast cancer cells supports two main conclusions. First, our approach to model construction can be used to build reasonably accurate quantitative predictors of pathway responses to combinatorial drug perturbatio
What problem does this paper attempt to address?
-
Biphasic Mathematical Model of Cell–Drug Interaction That Separates Target-Specific and Off-Target Inhibition and Suggests Potent Targeted Drug Combinations for Multi-Driver Colorectal Cancer Cells
Jinyan Shen,Li Li,Tao Yang,Paul S. Cohen,Gongqin Sun
DOI: https://doi.org/10.3390/cancers12020436
2020-02-13
Cancers
Abstract:Quantifying the response of cancer cells to a drug, and understanding the mechanistic basis of the response, are the cornerstones for anti-cancer drug discovery. Classical single target-based IC50 measurements are inadequate at describing cancer cell responses to targeted drugs. In this study, based on an analysis of targeted inhibition of colorectal cancer cell lines, we develop a new biphasic mathematical model that accurately describes the cell–drug response. The model describes the drug response using three kinetic parameters: ratio of target-specific inhibition, F1, potency of target-specific inhibition, Kd1, and potency of off-target toxicity, Kd2. Determination of these kinetic parameters also provides a mechanistic basis for predicting effective combination targeted therapy for multi-driver cancer cells. The experiments confirmed that a combination of inhibitors, each blocking a driver pathway and having a distinct target-specific effect, resulted in a potent and synergistic blockade of cell viability, improving potency over mono-agent treatment by one to two orders of magnitude. We further demonstrate that mono-driver cancer cells represent a special scenario in which F1 becomes nearly 100%, and the drug response becomes monophasic. Application of this model to the responses of >400 cell lines to kinase inhibitor dasatinib revealed that the ratio of biphasic versus monophasic responses is about 4:1. This study develops a new mathematical model of quantifying cancer cell response to targeted therapy, and suggests a new framework for developing rational combination targeted therapy for colorectal and other multi-driver cancers.
oncology
-
Model-based Analysis of Response and Resistance Factors of Cetuximab Treatment in Gastric Cancer Cell Lines
Elba Raimundez,Simone Keller,Gwen Zwingenberger,Karolin Ebert,Sabine Hug,Fabian J. Theis,Dieter Maier,Birgit Luber,Jan Hasenauer
DOI: https://doi.org/10.1371/journal.pcbi.1007147
2020-01-01
PLoS Computational Biology
Abstract:Targeted cancer therapies are powerful alternatives to chemotherapies or can be used complementary to these. Yet, the response to targeted treatments depends on a variety of factors, including mutations and expression levels, and therefore their outcome is difficult to predict. Here, we develop a mechanistic model of gastric cancer to study response and resistance factors for cetuximab treatment. The model captures the EGFR, ERK and AKT signaling pathways in two gastric cancer cell lines with different mutation patterns. We train the model using a comprehensive selection of time and dose response measurements, and provide an assessment of parameter and prediction uncertainties. We demonstrate that the proposed model facilitates the identification of causal differences between the cell lines. Furthermore, our study shows that the model provides predictions for the responses to different perturbations, such as knockdown and knockout experiments. Among other results, the model predicted the effect of MET mutations on cetuximab sensitivity. These predictive capabilities render the model a basis for the assessment of gastric cancer signaling and possibly for the development and discovery of predictive biomarkers.
-
Causal Models, Prediction, and Extrapolation in Cell Line Perturbation Experiments
James P. Long,Yumeng Yang,Kim-Anh Do
DOI: https://doi.org/10.48550/arXiv.2207.09991
2022-07-20
Abstract:In cell line perturbation experiments, a collection of cells is perturbed with external agents (e.g. drugs) and responses such as protein expression measured. Due to cost constraints, only a small fraction of all possible perturbations can be tested in vitro. This has led to the development of computational (in silico) models which can predict cellular responses to perturbations. Perturbations with clinically interesting predicted responses can be prioritized for in vitro testing. In this work, we compare causal and non-causal regression models for perturbation response prediction in a Melanoma cancer cell line. The current best performing method on this data set is Cellbox which models how proteins causally effect each other using a system of ordinary differential equations (ODEs). We derive a closed form solution to the Cellbox system of ODEs in the linear case. These analytic results facilitate comparison of Cellbox to regression approaches. We show that causal models such as Cellbox, while requiring more assumptions, enable extrapolation in ways that non-causal regression models cannot. For example, causal models can predict responses for never before tested drugs. We illustrate these strengths and weaknesses in simulations. In an application to the Melanoma cell line data, we find that regression models outperform the Cellbox causal model.
Applications,Quantitative Methods
-
Perturbation Biology: Inferring Signaling Networks in Cellular Systems
Evan J. Molinelli,Anil Korkut,Weiqing Wang,Martin L. Miller,Nicholas P. Gauthier,Xiaohong Jing,Poorvi Kaushik,Qin He,Gordon Mills,David B. Solit,Christine A. Pratilas,Martin Weigt,Alfredo Braunstein,Andrea Pagnani,Riccardo Zecchina,Chris Sander
DOI: https://doi.org/10.1371/journal.pcbi.1003290
2013-12-19
PLoS Computational Biology
Abstract:We present a powerful experimental-computational technology for inferring network models that predict the response of cells to perturbations, and that may be useful in the design of combinatorial therapy against cancer. The experiments are systematic series of perturbations of cancer cell lines by targeted drugs, singly or in combination. The response to perturbation is quantified in terms of relative changes in the measured levels of proteins, phospho-proteins and cellular phenotypes such as viability. Computational network models are derived de novo, i.e., without prior knowledge of signaling pathways, and are based on simple non-linear differential equations. The prohibitively large solution space of all possible network models is explored efficiently using a probabilistic algorithm, Belief Propagation (BP), which is three orders of magnitude faster than standard Monte Carlo methods. Explicit executable models are derived for a set of perturbation experiments in SKMEL-133 melanoma cell lines, which are resistant to the therapeutically important inhibitor of RAF kinase. The resulting network models reproduce and extend known pathway biology. They empower potential discoveries of new molecular interactions and predict efficacious novel drug perturbations, such as the inhibition of PLK1, which is verified experimentally. This technology is suitable for application to larger systems in diverse areas of molecular biology.
biochemical research methods,mathematical & computational biology
-
Predicting cell morphological responses to perturbations using generative modeling
A. Palma,Fabian J. Theis,Mohammad Lotfollahi
DOI: https://doi.org/10.1101/2023.07.17.549216
2023-01-01
Abstract:Abstract Advancements in high-throughput screening have enabled the exploration of rich phenotypic readouts like high-content microscopy, expediting drug target identification and mode of action studies. However, scaling these experiments to the vast space of drug or genetic manipulations poses challenges, as only a small subset of compounds show activity in screenings. Despite being widely used in various applications, machine learning methods have not shown a reliable ability to extrapolate predictions to scenarios involving unseen phenomena, specifically transforming an unseen control cell image into a desired perturbation. We present a generative model, the IMage Perturbation Autoencoder (IMPA), which predicts cellular morphological effects of chemical and genetic perturbations using untreated cells as input. IMPA learns perturbation-specific styles from generalized embeddings and generates counterfactual treatment response predictions in control cells. We demonstrate IMPA can predict morphological changes caused by small molecule perturbations on breast cancer cells. Additionally, we test IMPA on the unseen drug effect prediction task, showing improved performance over state-of-the-art generative models when compounds are structurally related to the training set. Finally, generalizability and capability to predict more subtle effects are showcased through its application to large microscopy datasets with hundreds of genetic perturbations on U2OS cells. We envision IMPA to become a valuable tool in computational microscopy for aiding phenotypic drug discovery, facilitating navigation of the perturbation space, and rational experimental design.
-
Abstract 4921: Dynamic and quantitative modeling of drug effect
Xiangfang Li,Lijun Qian,Michael Bittner,Edward Dougherty
DOI: https://doi.org/10.1158/1538-7445.am2011-4921
2011-04-15
Cellular and Molecular Biology
Abstract:Abstract The current drug development is a very expensive and prolonged process with high attrition rate, especially in the oncology drug development. New tools with dynamic and quantitative modeling and experimental support are needed to accelerate the drug discovery process and increase productivity. The process begins with building a quantitative model of biological regulatory system based on the extensive data-mining from literature and experiments. By incorporating the preclinical PK/PD data relevant to the target system, a new computational model under drug perturbation can be built. Through iterative process of hypotheses generating and experimental testing, such models can be fine-tuned. Consequences of particular perturbations, such as optimal dosage and treatment schedules, optimal drug target, and combination therapy can be simulated in time course using such models. A better understanding of the biological system will be gained and better therapeutic interventions suggestions can be provided. In this study, we use NF-κB pathway as an example study considering NF-κB pathway has become a focal point for intense drug discovery and development efforts. Dynamic drug perturbation is incorporated in the model stem from Hoffmann et al. (science 2002 Nov 8;298) through hybrid system theory to link dose – concentration relationships (PK) and concentration – effect relationships (PD). This approach facilitates the description and prediction of the time course of drug effects resulting from a certain dose regimen. Extensive simulations of the proposed model are performed in MATLAB/SIMULINK. An important observation is that there exists a “sweet spot” for certain dosage and frequency arrangements given the totaled delivered drug. The perturbation effect for different targets is simulated and compared in this model. However, the specificity of some drugs to inhibit several of these components of the NF-κB pathway is one of the concerns. For example, if we block the degradation pathway of IκB by using proteasome inhibitor (such as Bortezomib) to inhibit ubiquitination of IκB by the 26S protesome. The side effects are unacceptable when the drug is effective. Hence, we design combination therapy to induce a acceptable combined effect and using lower doses for each drugs. We observe that some combinations of the drugs with non-overlapping toxicities might provide enormous benefit. While such dynamical modeling approaches are still in their infancy, advancing techniques of the quantitative single live cell measurement of the protein processes will provide us more accurate parameters and help us to test and confirm some observations. This will allow a better understanding of the kinetics and function of mechanisms within regulatory networks. The authors believe that appropriate models can help establishing a mechanistic understanding of the disease and of drug action instead of the traditional “black-box” approach in drug discovery. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4921. doi:10.1158/1538-7445.AM2011-4921
cell biology,biochemistry & molecular biology
-
Quantifying assays: A Modeling tale of variability in cancer therapeutics assessed on cancer cells
Roumen Anguelov,G Manjunath,Avulundiah E Phiri,Trevor T Nyakudya,Priyesh Bipath,June C Serem,Yvette N Hlophe
DOI: https://doi.org/10.48550/arXiv.2207.08449
2022-07-18
Abstract:Inhibiting a signalling pathway concerns controlling the cellular processes of a cancer cell's viability, cell division, and death. Assay protocols created to see if the molecular structures of the drugs being tested have the desired inhibition qualities often show great variability across experiments, and it is imperative to diminish the effects of such variability while inferences are drawn. In this paper we propose the study of experimental data through the lenses of a mathematical model depicting the inhibition mechanism and the activation-inhibition dynamics. The method is exemplified through assay data obtained from the study of inhibition of the CXCL12/CXCR4 activation axis for the melanoma cells. To mitigate the effects of the variability of the data on the cell viability measurement, the cell viability is theoretically constructed as a function of time depending on several parameters. The values of these parameters are estimated by using the experimental data. Deriving approximation for the cell viability in a theoretically pre-determined form has the advantages of (i) being less sensitive to data variability (ii) the estimated values of the parameters are interpreted directly in the biological processes, (iii) the amount of variability explained via the approximation validates the quality of the model, (iv) with the data integrated into the model one can derive a more complete view over the whole process. These advantages are demonstrated in the step-by-step implementation of the outlined approach.
Quantitative Methods,Dynamical Systems,Cell Behavior
-
Systems Perturbation Analysis of a Large Scale Signal Transduction Model Reveals Potentially Influential Candidates for Cancer Therapeutics
Bhanwar Lal Puniya,Laura Allen,Colleen Hochfelder,Mahbubul Majumder,Tomáš Helikar
DOI: https://doi.org/10.48550/arXiv.1511.03337
2015-11-11
Abstract:Dysregulation in signal transduction pathways can lead to a variety of complex disorders, including cancer. Computational approaches such as network analysis are important tools to understand system dynamics as well as to identify critical components that could be further explored as therapeutic targets. Here, we performed perturbation analysis of a large-scale signal transduction model in extracellular environments that stimulate cell death, growth, motility, and quiescence. Each of the model's components was perturbed under both loss-of-function and gain-of-function mutations. We identified the most and least influential components based on the magnitude of their influence on the rest of the system. Based on the premise that the most influential components might serve as better drug targets, we characterized them for biological functions, housekeeping genes, essential genes, and druggable proteins. Moreover, known cancer drug targets were also classified in influential components based on the affected components in the network. Additionally, the systemic perturbation analysis of the model revealed a network motif of most influential components which affect each other. Furthermore, our analysis predicted novel combinations of cancer drug targets with various effects on other most influential components. We found that the combinatorial perturbation consisting of PI3K inactivation and overactivation of IP3R1 can lead to increased activity levels of apoptosis-related components and tumor suppressor genes, suggesting that this combinatorial perturbation may lead to a better target for decreasing cell proliferation and inducing apoptosis. Lastly, our results suggest that systematic perturbation analyses of large-scale computational models may serve as an approach to prioritize and assess signal transduction components in order to identify novel drug targets in complex disorders.
Molecular Networks
-
Mechanistic modeling of cell viability assays with in silico lineage tracing
Arnab Mutsuddy,Jonah R Huggins,Aurore Amrit,Cemal Erdem,Jon C Calhoun,Marc R Birtwistle
DOI: https://doi.org/10.1101/2024.08.23.609433
2024-08-26
Abstract:Data from cell viability assays, which measure cumulative division and death events in a population and reflect substantial cellular heterogeneity, are widely available. However, interpreting such data with mechanistic computational models is hindered because direct model/data comparison is often muddled. We developed an algorithm that tracks simulated division and death events in mechanistically detailed single-cell lineages to enable such a model/data comparison and suggest causes of cell-cell drug response variability. Using our previously developed model of mammalian single-cell proliferation and death signaling, we simulated drug dose response experiments for four targeted anti-cancer drugs (alpelisib, neratinib, trametinib and palbociclib) and compared them to experimental data. Simulations are consistent with data for strong growth inhibition by trametinib (MEK inhibitor) and overall lack of efficacy for alpelisib (PI-3K inhibitor), but are inconsistent with data for palbociclib (CDK4/6 inhibitor) and neratinib (EGFR inhibitor). Model/data inconsistencies suggest (i) the importance of CDK4/6 for driving the cell cycle may be overestimated, and (ii) that the cellular balance between basal (tonic) and ligand-induced signaling is a critical determinant of receptor inhibitor response. Simulations show subpopulations of rapidly and slowly dividing cells in both control and drug-treated conditions. Variations in mother cells prior to drug treatment all impinging on ERK pathway activity are associated with the rapidly dividing phenotype and trametinib resistance. This work lays a foundation for the application of mechanistic modeling to large-scale cell viability assay datasets and better understanding determinants of cellular heterogeneity in drug response.
Systems Biology
-
A mechanistic pan-cancer pathway model informed by multi-omics data interprets stochastic cell fate responses to drugs and mitogens
Mehdi Bouhaddou,Anne Marie Barrette,Alan D. Stern,Rick J. Koch,Matthew S. DiStefano,Eric A. Riesel,Luis C. Santos,Annie L. Tan,Alex E. Mertz,Marc R. Birtwistle
DOI: https://doi.org/10.1371/journal.pcbi.1005985
2018-03-26
PLoS Computational Biology
Abstract:Most cancer cells harbor multiple drivers whose epistasis and interactions with expression context clouds drug and drug combination sensitivity prediction. We constructed a mechanistic computational model that is context-tailored by omics data to capture regulation of stochastic proliferation and death by pan-cancer driver pathways. Simulations and experiments explore how the coordinated dynamics of RAF/MEK/ERK and PI-3K/AKT kinase activities in response to synergistic mitogen or drug combinations control cell fate in a specific cellular context. In this MCF10A cell context, simulations suggest that synergistic ERK and AKT inhibitor-induced death is likely mediated by BIM rather than BAD, which is supported by prior experimental studies. AKT dynamics explain S-phase entry synergy between EGF and insulin, but simulations suggest that stochastic ERK, and not AKT, dynamics seem to drive cell-to-cell proliferation variability, which in simulations is predictable from pre-stimulus fluctuations in C-Raf/B-Raf levels. Simulations suggest MEK alteration negligibly influences transformation, consistent with clinical data. Tailoring the model to an alternate cell expression and mutation context, a glioma cell line, allows prediction of increased sensitivity of cell death to AKT inhibition. Our model mechanistically interprets context-specific landscapes between driver pathways and cell fates, providing a framework for designing more rational cancer combination therapy.
biochemical research methods,mathematical & computational biology
-
Computational Modeling of Drug Response Identifies Mutant-Specific Constraints for Dosing panRAF and MEK Inhibitors in Melanoma
Andrew Goetz,Frances Shanahan,Logan Brooks,Eva Lin,Rana Mroue,Darlene Dela Cruz,Thomas Hunsaker,Bartosz Czech,Purushottam Dixit,Udi Segal,Scott Martin,Scott A. Foster,Luca Gerosa
DOI: https://doi.org/10.3390/cancers16162914
2024-08-24
Cancers
Abstract:Purpose: This study explores the potential of pre-clinical in vitro cell line response data and computational modeling in identifying the optimal dosage requirements of pan-RAF (Belvarafenib) and MEK (Cobimetinib) inhibitors in melanoma treatment. Our research is motivated by the critical role of drug combinations in enhancing anti-cancer responses and the need to close the knowledge gap around selecting effective dosing strategies to maximize their potential. Results: In a drug combination screen of 43 melanoma cell lines, we identified specific dosage landscapes of panRAF and MEK inhibitors for NRAS vs. BRAF mutant melanomas. Both experienced benefits, but with a notably more synergistic and narrow dosage range for NRAS mutant melanoma (mean Bliss score of 0.27 in NRAS vs. 0.1 in BRAF mutants). Computational modeling and follow-up molecular experiments attributed the difference to a mechanism of adaptive resistance by negative feedback. We validated the in vivo translatability of in vitro dose–response maps by predicting tumor growth in xenografts with high accuracy in capturing cytostatic and cytotoxic responses. We analyzed the pharmacokinetic and tumor growth data from Phase 1 clinical trials of Belvarafenib with Cobimetinib to show that the synergy requirement imposes stricter precision dose constraints in NRAS mutant melanoma patients. Conclusion: Leveraging pre-clinical data and computational modeling, our approach proposes dosage strategies that can optimize synergy in drug combinations, while also bringing forth the real-world challenges of staying within a precise dose range. Overall, this work presents a framework to aid dose selection in drug combinations.
oncology
-
Abstract 2723: Model-based cancer therapy selection by linking tumor vulnerabilities to drug mechanism
Szabolcs Tarapcsak,Yi Qiao,Xiaomeng Huang,Tony Di Sera,Matthew H. Bailey,Bryan E. Welm,Alana L. Welm,Gabor T. Marth
DOI: https://doi.org/10.1158/1538-7445.am2022-2723
IF: 11.2
2022-06-17
Cancer Research
Abstract:Selection of the treatments most likely to combat a patient's tumor is a central aim of precision oncology. We are currently developing a functional precision oncology program at the University of Utah and Huntsman Cancer Institute that combines the multi-omic characterization of a patient's tumor with the functional screening of relevant drug candidates in patient-derived organoid tumor models to identify which drugs are uniquely capable of combating the patient's cancer. Here we present our model-based approach for utilizing the multi-omic tumor data to computationally predict the patient's response to each drug. In this approach, we first identify the genomic and transcriptomic vulnerabilities of the tumor as genes harboring somatic DNA mutations or copy number changes (from paired tumor/normal WGS/WES DNA sequencing data), as well as genes whose expression levels have been significantly altered in the tumor (based on bulk or single-cell RNA sequencing data). Second, using targeting information from drug-gene interaction databases (DGIdb) we compile a list of genes targeted by each drug relevant in the patient's treatment. Third, we utilize gene interaction database knowledge (KEGG) to construct a gene interaction graph to link each drug's gene targets with all of the tumor's vulnerability genes. We then identify all gene interaction paths connecting specific drug target genes with specific vulnerability genes; and score each path and combine all path-specific scores for the target-vulnerability gene pair. Subsequently, we further combine all pairwise scores across all drug target genes and all tumor vulnerability genes and determine the statistical significance of the resulting score by sampling the background distribution of such scores across random drugs, target genes, and vulnerability genes. We have applied our algorithm to predict drug response in advanced breast cancer patients as well as using publicly available tumor cell line data (GDSC2). We have found a high level of concordance between our computational prediction and organoid/cell line screening results, clearly separating sensitive and non-sensitive models. Using Bayesian probability, we compare the drug-specific score distributions of sensitive and non-sensitive models and are able to identify sensitive cell lines/patients with high accuracy. By encapsulating available information on direct gene-gene interactions, the drug's direct gene targets and the collected omic vulnerabilities of the tumor, our model is not only capable of predicting sensitivity to both targeted and chemotherapy agents, but can also provide a mechanistic understanding for the targeting of the tumor. Our approach will be validated and fine-tuned on a large cohort of breast and brain cancer patients as well as patient PDX models, interrogating gene-target interactions to identify novel relevant target genes/pathways. Citation Format: Szabolcs Tarapcsak, Yi Qiao, Xiaomeng Huang, Tony Di Sera, Matthew H. Bailey, Bryan E. Welm, Alana L. Welm, Gabor T. Marth. Model-based cancer therapy selection by linking tumor vulnerabilities to drug mechanism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl) nr 2723.
oncology
-
Patient-derived models of acquired resistance can identify effective drug combinations for cancer
Adam S Crystal,Alice T Shaw,Lecia V Sequist,Luc Friboulet,Matthew J Niederst,Elizabeth L Lockerman,Rosa L Frias,Justin F Gainor,Arnaud Amzallag,Patricia Greninger,Dana Lee,Anuj Kalsy,Maria Gomez-Caraballo,Leila Elamine,Emily Howe,Wooyoung Hur,Eugene Lifshits,Hayley E Robinson,Ryohei Katayama,Anthony C Faber,Mark M Awad,Sridhar Ramaswamy,Mari Mino-Kenudson,A John Iafrate,Cyril H Benes,Jeffrey A Engelman,Adam S. Crystal,Alice T. Shaw,Lecia V. Sequist,Matthew J. Niederst,Elizabeth L. Lockerman,Rosa L. Frias,Justin F. Gainor,Hayley E. Robinson,Anthony C. Faber,Mark M. Awad,A. John Iafrate,Cyril H. Benes,Jeffrey A. Engelman
DOI: https://doi.org/10.1126/science.1254721
IF: 56.9
2014-12-19
Science
Abstract:Targeted cancer therapies have produced substantial clinical responses, but most tumors develop resistance to these drugs. Here, we describe a pharmacogenomic platform that facilitates rapid discovery of drug combinations that can overcome resistance. We established cell culture models derived from biopsy samples of lung cancer patients whose disease had progressed while on treatment with epidermal growth factor receptor (EGFR) or anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors and then subjected these cells to genetic analyses and a pharmacological screen. Multiple effective drug combinations were identified. For example, the combination of ALK and MAPK kinase (MEK) inhibitors was active in an ALK -positive resistant tumor that had developed a MAP2K1 activating mutation, and the combination of EGFR and fibroblast growth factor receptor (FGFR) inhibitors was active in an EGFR mutant resistant cancer with a mutation in FGFR3 . Combined ALK and SRC (pp60c-src) inhibition was effective in several ALK-driven patient-derived models, a result not predicted by genetic analysis alone. With further refinements, this strategy could help direct therapeutic choices for individual patients.
multidisciplinary sciences
-
Bifurcation-based approach reveals synergism and optimal combinatorial perturbation
Yanwei Liu,Shanshan Li,Zengrong Liu,Ruiqi Wang
DOI: https://doi.org/10.1007/s10867-016-9414-7
Abstract:Cells accomplish the process of fate decisions and form terminal lineages through a series of binary choices in which cells switch stable states from one branch to another as the interacting strengths of regulatory factors continuously vary. Various combinatorial effects may occur because almost all regulatory processes are managed in a combinatorial fashion. Combinatorial regulation is crucial for cell fate decisions because it may effectively integrate many different signaling pathways to meet the higher regulation demand during cell development. However, whether the contribution of combinatorial regulation to the state transition is better than that of a single one and if so, what the optimal combination strategy is, seem to be significant issue from the point of view of both biology and mathematics. Using the approaches of combinatorial perturbations and bifurcation analysis, we provide a general framework for the quantitative analysis of synergism in molecular networks. Different from the known methods, the bifurcation-based approach depends only on stable state responses to stimuli because the state transition induced by combinatorial perturbations occurs between stable states. More importantly, an optimal combinatorial perturbation strategy can be determined by investigating the relationship between the bifurcation curve of a synergistic perturbation pair and the level set of a specific objective function. The approach is applied to two models, i.e., a theoretical multistable decision model and a biologically realistic CREB model, to show its validity, although the approach holds for a general class of biological systems.
-
Converting networks to predictive logic models from perturbation signalling data with CellNOpt
Enio Gjerga,Panuwat Trairatphisan,Attila Gabor,Hermann Koch,Celine Chevalier,Franceco Ceccarelli,Aurelien Dugourd,Alexander Mitsos,Julio Saez-Rodriguez
DOI: https://doi.org/10.1093/bioinformatics/btaa561
IF: 5.8
2020-06-09
Bioinformatics
Abstract:Abstract Summary The molecular changes induced by perturbations such as drugs and ligands are highly informative of the intracellular wiring. Our capacity to generate large datasets is increasing steadily. A useful way to extract mechanistic insight from the data is by integrating them with a prior knowledge network of signalling to obtain dynamic models. CellNOpt is a collection of Bioconductor R packages for building logic models from perturbation data and prior knowledge of signalling networks. We have recently developed new components and refined the existing ones to keep up with the computational demand of increasingly large datasets, including (i) an efficient integer linear programming, (ii) a probabilistic logic implementation for semi-quantitative datasets, (iii) the integration of a stochastic Boolean simulator, (iv) a tool to identify missing links, (v) systematic post-hoc analyses and (vi) an R-Shiny tool to run CellNOpt interactively. Availability and implementation R-package(s): https://github.com/saezlab/cellnopt. Supplementary information Supplementary data are available at Bioinformatics online.
biochemical research methods,biotechnology & applied microbiology,mathematical & computational biology
-
Reconstruction of cellular signal transduction networks using perturbation assays and linear programming
Bettina Knapp,Lars Kaderali
DOI: https://doi.org/10.1371/journal.pone.0069220
IF: 3.7
2013-07-30
PLoS ONE
Abstract:Perturbation experiments for example using RNA interference (RNAi) offer an attractive way to elucidate gene function in a high throughput fashion. The placement of hit genes in their functional context and the inference of underlying networks from such data, however, are challenging tasks. One of the problems in network inference is the exponential number of possible network topologies for a given number of genes. Here, we introduce a novel mathematical approach to address this question. We formulate network inference as a linear optimization problem, which can be solved efficiently even for large-scale systems. We use simulated data to evaluate our approach, and show improved performance in particular on larger networks over state-of-the art methods. We achieve increased sensitivity and specificity, as well as a significant reduction in computing time. Furthermore, we show superior performance on noisy data. We then apply our approach to study the intracellular signaling of human primary nave CD4(+) T-cells, as well as ErbB signaling in trastuzumab resistant breast cancer cells. In both cases, our approach recovers known interactions and points to additional relevant processes. In ErbB signaling, our results predict an important role of negative and positive feedback in controlling the cell cycle progression.
-
Characterizing heterogeneous single-cell dose responses computationally and experimentally using threshold inhibition surfaces and dose-titration assays
Patrick C. Kinnunen,Brock A. Humphries,Gary D. Luker,Kathryn E. Luker,Jennifer J. Linderman
DOI: https://doi.org/10.1038/s41540-024-00369-x
2024-04-18
npj Systems Biology and Applications
Abstract:Abstract Single cancer cells within a tumor exhibit variable levels of resistance to drugs, ultimately leading to treatment failures. While tumor heterogeneity is recognized as a major obstacle to cancer therapy, standard dose-response measurements for the potency of targeted kinase inhibitors aggregate populations of cells, obscuring intercellular variations in responses. In this work, we develop an analytical and experimental framework to quantify and model dose responses of individual cancer cells to drugs. We first explore the connection between population and single-cell dose responses using a computational model, revealing that multiple heterogeneous populations can yield nearly identical population dose responses. We demonstrate that a single-cell analysis method, which we term a threshold inhibition surface, can differentiate among these populations. To demonstrate the applicability of this method, we develop a dose-titration assay to measure dose responses in single cells. We apply this assay to breast cancer cells responding to phosphatidylinositol-3-kinase inhibition (PI3Ki), using clinically relevant PI3Kis on breast cancer cell lines expressing fluorescent biosensors for kinase activity. We demonstrate that MCF-7 breast cancer cells exhibit heterogeneous dose responses with some cells requiring over ten-fold higher concentrations than the population average to achieve inhibition. Our work reimagines dose-response relationships for cancer drugs in an emerging paradigm of single-cell tumor heterogeneity.
mathematical & computational biology
-
Predicting Drug Combination Index and Simulating the Network-Regulation Dynamics by Mathematical Modeling of Drug-Targeted EGFR-ERK Signaling Pathway
Lu Huang,Yuyang Jiang,Yuzong Chen
DOI: https://doi.org/10.1038/srep40752
IF: 4.6
2017-01-01
Scientific Reports
Abstract:Synergistic drug combinations enable enhanced therapeutics. Their discovery typically involves the measurement and assessment of drug combination index (CI), which can be facilitated by the development and applications of in-silico CI predictive tools. In this work, we developed and tested the ability of a mathematical model of drug-targeted EGFR-ERK pathway in predicting CIs and in analyzing multiple synergistic drug combinations against observations. Our mathematical model was validated against the literature reported signaling, drug response dynamics, and EGFR-MEK drug combination effect. The predicted CIs and combination therapeutic effects of the EGFR-BRaf, BRaf-MEK, FTI-MEK, and FTI-BRaf inhibitor combinations showed consistent synergism. Our results suggest that existing pathway models may be potentially extended for developing drug-targeted pathway models to predict drug combination CI values, isobolograms, and drug-response surfaces as well as to analyze the dynamics of individual and combinations of drugs. With our model, the efficacy of potential drug combinations can be predicted. Our method complements the developed in-silico methods (e.g. the chemogenomic profile and the statistically-inferenced network models) by predicting drug combination effects from the perspectives of pathway dynamics using experimental or validated molecular kinetic constants, thereby facilitating the collective prediction of drug combination effects in diverse ranges of disease systems.
-
Abstract A023: Integrating mathematical modeling and in vitro experiments to measure ecological interactions in cancer
Maximilian A. R. Strobl,Dagim S. Tadele,Jeffrey Maltas,Rowan Barker-Clarke,Mina Dinh,Jacob Scott
DOI: https://doi.org/10.1158/1538-7445.canevol23-a023
IF: 11.2
2024-02-02
Cancer Research
Abstract:Integrating mathematical modeling and in vitro experiments to measure ecological interactions in cancer Maximilian Strobl, Dagim Tadele, Jeffrey Maltas, Rowan Barker-Clarke, Mina Dinh, Jacob Scott Over the past two decades it has become clear that tumours are complex and evolving ecosystems, in which different cancer cell populations interact with each other and with their non-tumour microenvironment. This understanding has given rise to the intriguing idea that we might be able to leverage these interactions to slow, or even revert, the expansion of resistant cells. In so-called "Adaptive Therapy" (AT) treatment is dynamically reduced to maintain the tumour burden tolerable whilst also preserving a pool of drug-sensitive cells which competitively suppress resistant cells. However, how strong and how common is this suppression, and how do we stratify patients who might benefit from AT from those who should receive standard-of-care continuous therapy? In this contribution, we present development of methodology and initial experimental results aimed at measuring the distribution of ecological interactions between drug-sensitive and resistant cells. Previously, we pioneered an assay with which to quantify ecological interactions in cancer in vitro. In the "Game Assay", the two populations are co-cultured at a range of initial ratios in 2-D, and we record how the growth rate of each population is modulated by the relative abundance of the other. In the current study, we begin by using an agent-based mathematical model to characterize the accuracy and precision of this assay and how we may improve these by modifying different aspects of the experimental design (number of replicates, number of proportions) and analysis (regression technique, regression window). This shows that the choice of seeding ratios is important in being able to reliably identify interactions and we discuss guidelines for assay design. Upon identifying an interaction, a key next question is the underlying mechanism. Next, we thus use the model to investigate the degree to which we can use the assay to distinguish between local (e.g. contact inhibition) and non-local interactions (e.g. paracrine signaling). To conclude, we present preliminary experimental data in which we have used our optimized protocol to quantify the distribution of interactions between Osimertinib sensitive and resistant NSCLC cells (PC9). Our data show that while suppressive interactions do occur, they vary in their strength between different resistant lineages and can be attenuated in a frequency-dependent manner. Overall, we demonstrate how mathematical models can help in the design and interpretation of experimental assays and we contribute towards a more systematic and quantitative understanding of tumor ecology as a foundation for adaptive therapy. Citation Format: Maximilian A. R. Strobl, Dagim S. Tadele, Jeffrey Maltas, Rowan Barker-Clarke, Mina Dinh, Jacob Scott. Integrating mathematical modeling and in vitro experiments to measure ecological interactions in cancer [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Translating Cancer Evolution and Data Science: The Next Frontier; 2023 Dec 3-6; Boston, Massachusetts. Philadelphia (PA): AACR; Cancer Res 2024;84(3 Suppl_2) nr A023.
oncology