AUTHOR=Sivakumar Nikita , Mura Cameron , Peirce Shayn M. TITLE=Innovations in integrating machine learning and agent-based modeling of biomedical systems JOURNAL=Frontiers in Systems Biology VOLUME=2 YEAR=2022 URL=https://www.frontiersin.org/journals/systems-biology/articles/10.3389/fsysb.2022.959665 DOI=10.3389/fsysb.2022.959665 ISSN=2674-0702 ABSTRACT=

Agent-based modeling (ABM) is a well-established computational paradigm for simulating complex systems in terms of the interactions between individual entities that comprise the system’s population. Machine learning (ML) refers to computational approaches whereby algorithms use statistical methods to “learn” from data on their own, i.e., without imposing any a priori model/theory onto a system or its behavior. Biological systems—ranging from molecules, to cells, to entire organisms, to whole populations and even ecosystems—consist of vast numbers of discrete entities, governed by complex webs of interactions that span various spatiotemporal scales and exhibit nonlinearity, stochasticity, and variable degrees of coupling between entities. For these reasons, the macroscopic properties and collective dynamics of biological systems are generally difficult to accurately model or predict via continuum modeling techniques and mean-field formalisms. ABM takes a “bottom-up” approach that obviates common difficulties of other modeling approaches by enabling one to relatively easily create (or at least propose, for testing) a set of well-defined “rules” to be applied to the individual entities (agents) in a system. Quantitatively evaluating a system and propagating its state over a series of discrete time-steps effectively simulates the system, allowing various observables to be computed and the system’s properties to be analyzed. Because the rules that govern an ABM can be difficult to abstract and formulate from experimental data, at least in an unbiased way, there is a uniquely synergistic opportunity to employ ML to help infer optimal, system-specific ABM rules. Once such rule-sets are devised, running ABM calculations can generate a wealth of data, and ML can be applied in that context too—for example, to generate statistical measures that accurately and meaningfully describe the stochastic outputs of a system and its properties. As an example of synergy in the other direction (from ABM to ML), ABM simulations can generate plausible (realistic) datasets for training ML algorithms (e.g., for regularization, to mitigate overfitting). In these ways, one can envision a variety of synergistic ABM⇄ML loops. After introducing some basic ideas about ABMs and ML, and their limitations, this Review describes examples of how ABM and ML have been integrated in diverse contexts, spanning spatial scales that include multicellular and tissue-scale biology to human population-level epidemiology. In so doing, we have used published studies as a guide to identify ML approaches that are well-suited to particular types of ABM applications, based on the scale of the biological system and the properties of the available data.