Appearance
This report is written by MaltSci based on the latest literature and research findings
How does computational modeling simulate biological processes?
Abstract
The advent of computational modeling has revolutionized biomedical research by providing a powerful framework to simulate and analyze complex biological processes. This approach allows researchers to bridge the gap between molecular interactions and organ dynamics, enhancing our understanding of disease mechanisms, drug development, and personalized medicine. The report reviews various computational modeling techniques, including agent-based modeling, molecular dynamics simulations, and multiscale modeling, each offering unique advantages for studying biological systems. Applications in simulating disease mechanisms, optimizing drug discovery, and facilitating personalized medicine are discussed, highlighting the potential of these models to improve therapeutic strategies. However, challenges such as data availability, model validation, and computational resource constraints remain significant hurdles. Future directions emphasize the integration of multi-omics data, advancements in computational power, and enhancing model interoperability to foster a more comprehensive understanding of biological phenomena. Overall, computational modeling stands as a transformative tool that not only supports hypothesis generation and experimental design but also paves the way for innovative solutions in healthcare.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 The Role of Computational Modeling in Biology
- 2.1 Overview of Computational Modeling Techniques
- 2.2 Importance of Modeling in Biological Research
- 3 Applications of Computational Modeling in Biomedical Research
- 3.1 Disease Mechanism Simulation
- 3.2 Drug Discovery and Development
- 3.3 Personalized Medicine Approaches
- 4 Methodologies in Computational Modeling
- 4.1 Statistical Mechanics and Thermodynamics
- 4.2 Machine Learning Applications
- 4.3 Agent-Based Modeling Techniques
- 5 Challenges and Limitations
- 5.1 Data Availability and Quality
- 5.2 Model Validation and Reproducibility
- 5.3 Computational Resource Constraints
- 6 Future Directions in Computational Modeling
- 6.1 Integrating Multi-Omics Data
- 6.2 Advancements in Computational Power
- 6.3 Enhancing Model Interoperability
- 7 Conclusion
1 Introduction
The advent of computational modeling has revolutionized the landscape of biomedical research, enabling researchers to simulate and analyze complex biological processes with unprecedented precision. As biological systems operate at multiple scales—from molecular interactions to organ dynamics—computational modeling provides a versatile framework to bridge these disparate levels of complexity. The integration of mathematical frameworks and computational algorithms facilitates the creation of models that not only replicate biological behaviors but also predict outcomes under varying conditions. This capability is particularly crucial in an era where the demand for innovative approaches to understand disease mechanisms, enhance drug development, and personalize medicine is ever-increasing.
The significance of computational modeling in biomedical research cannot be overstated. It serves as a powerful tool for hypothesis generation, experimental design, and data interpretation, ultimately contributing to a more profound understanding of biological phenomena. Traditional experimental approaches often face limitations in terms of scalability, ethical considerations, and the ability to capture the dynamic nature of biological systems. Computational models, on the other hand, allow for the exploration of "what if" scenarios, thereby providing insights that can guide experimental efforts and inform clinical applications[1][2]. For instance, biosimulation techniques are increasingly being utilized to characterize complex interactions within biological systems, leading to enhanced predictive capabilities in drug discovery and development[3].
The current state of computational modeling in biomedicine reflects a robust and rapidly evolving field. The proliferation of literature on the subject highlights a growing recognition of its importance, with over 58,000 entries related to modeling in biological contexts available on PubMed[4]. Recent advancements have led to the development of various modeling techniques, including molecular dynamics simulations, statistical mechanics frameworks, and agent-based modeling, each offering unique advantages for specific research questions[5][6]. The convergence of computational and experimental methodologies has fostered a synergistic relationship, accelerating the pace of discovery and innovation in biomedicine.
This report is structured to provide a comprehensive overview of the role of computational modeling in simulating biological processes. The first section will delve into the various computational modeling techniques employed in biological research, emphasizing their importance and applications. Subsequent sections will explore specific applications in biomedical research, including disease mechanism simulation, drug discovery and development, and personalized medicine approaches. Furthermore, we will examine the methodologies underpinning these models, highlighting statistical mechanics, machine learning, and agent-based modeling techniques. The challenges and limitations faced by researchers in this domain will also be addressed, including issues related to data availability, model validation, and computational resource constraints. Finally, we will discuss future directions in computational modeling, emphasizing the integration of multi-omics data, advancements in computational power, and the enhancement of model interoperability.
Through this exploration, we aim to elucidate the transformative impact of computational modeling on our understanding of biological processes and its potential to improve patient outcomes in healthcare. The insights gained from this report will not only underscore the critical role of computational approaches in contemporary biomedical research but also highlight the ongoing challenges and opportunities that lie ahead in this dynamic field.
2 The Role of Computational Modeling in Biology
2.1 Overview of Computational Modeling Techniques
Computational modeling plays a pivotal role in simulating biological processes, allowing researchers to study complex systems that would be difficult to analyze through traditional experimental methods. The methodologies employed in computational modeling can be categorized into various approaches, each tailored to address specific aspects of biological phenomena.
Agent-based modeling (ABM) is one prominent technique that utilizes individual agents to represent biological entities, enabling the simulation of their interactions within a defined environment. This method has been particularly effective in oncology, where it helps in understanding tumor dynamics and the behavior of cancer cells under various conditions. According to a review by Stephan et al. (2024), ABM and other computational approaches have been instrumental in improving diagnosis and understanding the behaviors of biological entities, despite challenges such as the need for holistic models that encompass multiple scales and levels of organization [6].
Another significant method is object-oriented computational modeling, which constructs abstractions of biological objects and simulates their interactions. Johnson et al. (2004) emphasize that this approach is particularly useful for simulating complex biochemical processes within cells, allowing researchers to observe emergent properties and gain insights into cellular behaviors [7].
Molecular simulations are also a key component of computational modeling, enabling the exploration of biological processes at the molecular level. Trovato and Fumagalli (2017) discuss how molecular simulations can replicate cellular compartments, providing insights into macromolecule diffusion, nuclear body formation, and genetic material processes [8]. These simulations often require simplifications to balance accuracy and computational feasibility, as highlighted by Dokholyan (2006), who notes that simplified models can effectively capture biological phenomena across varying scales [9].
Furthermore, the integration of multiscale modeling techniques allows for a comprehensive understanding of biological systems by linking molecular interactions to cellular behavior and beyond. Meier-Schellersheim et al. (2009) point out that combining different scales of experimental research with appropriate computational techniques enables a more nuanced exploration of biological systems [10].
The application of computational models extends to drug discovery and development, where they are employed to predict the effects of molecular interventions at the organism level. Kumar et al. (2006) assert that computational modeling is essential for transitioning biology from a descriptive to a predictive science, thereby enhancing the efficiency of therapeutic development [3].
In summary, computational modeling encompasses a variety of techniques, including agent-based modeling, object-oriented modeling, and molecular simulations, each contributing to the understanding of complex biological processes. These models not only facilitate the interpretation of experimental data but also enable researchers to formulate new hypotheses and design innovative experimental strategies. The ongoing advancements in computational methods continue to revolutionize the field of biology, offering unprecedented insights into the mechanisms that govern living organisms.
2.2 Importance of Modeling in Biological Research
Computational modeling plays a crucial role in simulating biological processes, offering insights that complement experimental approaches and enhancing our understanding of complex biological systems. This multifaceted approach involves various methodologies and models, each tailored to address specific biological questions across different scales.
At its core, computational modeling utilizes computer simulations to replicate and analyze biological phenomena, which is particularly significant in the context of understanding interactions among biological elements. For instance, in oncology, agent-based modeling (ABM) has been highlighted as a promising method to simulate tumor behavior and treatment responses, effectively capturing the dynamics of biological entities through computational techniques [6]. These models allow researchers to explore complex interactions that are often difficult to observe experimentally, thereby providing a deeper understanding of the underlying mechanisms of diseases.
The methodology of computational modeling encompasses a variety of approaches, including simplified molecular models and multiscale models. Simplified models, while less detailed than all-atom simulations, can effectively bridge the gap between different scales of biological phenomena. They have been shown to accurately reproduce essential biological behaviors, such as protein folding and aggregation, thus offering novel insights into processes that are not easily captured through traditional experimental methods [9]. Furthermore, the integration of simplified models with more detailed simulations can enhance the overall understanding of biological processes, allowing researchers to access a broader range of temporal and spatial scales [9].
Moreover, the use of computational models extends to simulating cellular processes under various conditions. For example, researchers have successfully modeled macromolecule diffusion and nuclear body formation, addressing challenges associated with simulating slow biological processes. The development of these models often requires careful consideration of structural simplifications and energetic descriptions to ensure accuracy while exploring complex cellular behaviors [8].
In addition to simulating specific biological processes, computational models facilitate hypothesis generation and testing. They provide a framework for comparing competing hypotheses, interpreting complex data, and investigating questions that are challenging to approach through experimental means [11]. This capability is particularly valuable in reproductive biology, where models have been employed to elucidate mechanisms governing germ line development across various organisms [11].
The importance of computational modeling in biological research cannot be overstated. It serves as a transformative tool that not only enhances the predictive capabilities of biological studies but also supports drug discovery and development by identifying potential therapeutic targets and assessing their effects in silico [3]. As the field of biology continues to evolve, the integration of computational methods will likely lead to more refined models that accurately reflect the complexities of biological systems, ultimately driving advancements in biomedical research and therapeutic strategies [1].
In conclusion, computational modeling stands as an indispensable component of modern biological research, enabling scientists to simulate and analyze biological processes with unprecedented precision. By bridging the gap between experimental observations and theoretical predictions, these models provide a comprehensive understanding of the intricate dynamics governing life at various scales, from molecular interactions to organism-level responses.
3 Applications of Computational Modeling in Biomedical Research
3.1 Disease Mechanism Simulation
Computational modeling has emerged as a pivotal tool in simulating biological processes, particularly within the realm of biomedical research. It offers various methodologies to explore and understand complex biological systems, bridging the gap between theoretical frameworks and experimental observations. The applications of computational modeling in simulating disease mechanisms are multifaceted and include several key approaches.
One significant aspect of computational modeling is its ability to integrate in silico techniques with experimental data, thus enhancing the predictive accuracy of biological systems. For instance, the application of molecular dynamics (MD) simulations allows researchers to study the behavior of biomolecules at atomic levels, providing insights into their structural, functional, and evolutionary characteristics over time. This approach is fundamental in understanding interactions at the molecular scale, which is crucial for assessing the properties of biomaterials and their interactions with biological environments [5].
Moreover, computational models can facilitate the simulation of disease mechanisms by employing agent-based modeling (ABM). This method allows for the exploration of interactions among biological entities, enabling the study of emergent behaviors within complex biological systems. ABM has been particularly relevant in oncology, where it helps improve the understanding of tumor dynamics and the interactions between cancer cells and their microenvironment [6]. The ability to simulate these interactions in a controlled environment aids in identifying potential therapeutic targets and predicting responses to treatments.
In addition, the development of multi-physics simulation platforms has further advanced the capability to model complex biological systems. These platforms can simulate various physiological processes, such as neuronal activity and perfusion, within realistic anatomical models. Such comprehensive simulations are essential for applications in device development, treatment planning, and safety assessments [12]. They allow researchers to visualize the intricate dynamics of biological systems, providing a holistic view of disease mechanisms.
Furthermore, the use of computational models in drug discovery is increasingly recognized as vital for translating biological insights into therapeutic interventions. By modeling cellular and tissue responses to pharmacological agents, researchers can predict the efficacy and safety of new drugs, thereby streamlining the drug development pipeline [3]. This predictive capability is crucial in addressing the challenges associated with late-stage drug failures and the rising costs of drug development.
Lastly, computational modeling supports personalized medicine by allowing the creation of individualized models that reflect the unique biological characteristics of patients. This approach enables the design of tailored therapeutic strategies that are more likely to succeed in treating specific disease mechanisms [13].
In summary, computational modeling serves as a powerful tool in simulating biological processes, particularly in understanding disease mechanisms. Its applications range from molecular dynamics and agent-based modeling to multi-physics simulations and personalized medicine, all contributing to a more nuanced understanding of biological systems and the development of effective therapeutic strategies. As computational methods continue to evolve, their integration into biomedical research promises to enhance our ability to tackle complex health challenges.
3.2 Drug Discovery and Development
Computational modeling plays a crucial role in simulating biological processes, particularly in the context of drug discovery and development. It encompasses a variety of techniques that allow researchers to understand complex biological systems, predict drug interactions, and identify potential therapeutic targets.
One of the primary applications of computational modeling in drug discovery is its ability to analyze cancer dynamics, as highlighted in melanoma research. In this area, computational modeling has been applied to discover new molecular targets that can lead to novel therapies and help overcome resistance to existing anticancer drugs. Melanoma, known for its aggressive nature and poor prognosis, serves as a compelling tumor model where these computational approaches can yield significant insights (Pennisi et al. 2016) [14].
Furthermore, computational drug discovery has become an effective strategy to accelerate and economize the drug development process. With advancements in computational methods such as molecular docking, pharmacophore modeling, and virtual screening, researchers can identify and validate drug targets, optimize lead compounds, and conduct preclinical tests more efficiently (Ou-Yang et al. 2012) [15]. These methods facilitate a comprehensive understanding of drug-target interactions, thereby streamlining the discovery process.
Recent developments have also emphasized the integration of high-throughput virtual screening and molecular dynamics simulations in identifying drug candidates. For instance, computational techniques have been successfully applied to various diseases, including breast cancer, where specific therapeutic targets such as estrogen receptors and HER2 are investigated. These modeling tools not only aid in the identification of drug candidates but also enhance the understanding of their binding mechanisms and kinetic profiles (Odunitan et al. 2024) [16].
Moreover, computational modeling has evolved to encompass the simulation of biological pathways and the prediction of drug efficacy. This includes modeling the interactions between different molecular pathways and the effects of drug cocktails, which can significantly improve therapeutic strategies (Yao et al. 2009) [17]. The ability to predict how different drugs might work together is particularly valuable in designing combination therapies for complex diseases.
The reliance on computational approaches has also been driven by the limitations of traditional experimental methods, particularly animal models, which are often costly and ethically contentious. Computational simulation models provide a viable alternative, enabling researchers to replicate human physiological and pathological processes with greater precision. This shift towards in silico methods not only enhances predictive accuracy but also aligns with ethical initiatives promoting humane research practices (Mittal et al. 2025) [1].
In conclusion, computational modeling serves as an indispensable tool in biomedical research, particularly in drug discovery and development. By simulating biological processes, it enables the identification of novel drug targets, optimizes therapeutic strategies, and ultimately contributes to the advancement of precision medicine. The integration of these advanced computational techniques into the drug development workflow is essential for addressing the complexities of modern pharmacotherapy and improving health outcomes.
3.3 Personalized Medicine Approaches
Computational modeling plays a critical role in simulating biological processes, particularly within the realm of biomedical research. It encompasses a variety of methodologies that enable the exploration of complex biological systems, enhancing our understanding and providing innovative approaches to personalized medicine.
Computational models, including in silico simulations, are employed to predict biological behavior under different conditions, bridging gaps in predictive accuracy and translational relevance. These models can reduce reliance on traditional animal models, which face ethical concerns and limitations in translational relevance to human biology. For instance, a systematic review by Mittal et al. (2025) highlights that computational simulation models support drug development pipelines, reduce late-stage failures, and enhance opportunities for personalized medicine by allowing researchers to simulate human physiological and pathological processes with greater precision[1].
In the context of personalized medicine, computational modeling provides a framework for creating individualized computational models that can inform treatment decisions tailored to specific patient profiles. This approach is exemplified by the use of mechanistic computational modeling to generate in silico clinical trials, which can be customized based on individual patient data and disease mechanisms. Such models facilitate the testing of hypotheses in a controlled environment, allowing researchers to explore the effects of different therapeutic interventions without the ethical and practical limitations of traditional experimental approaches[13].
Furthermore, molecular dynamics (MD) simulations are particularly relevant in understanding the behavior of biopolymers and biomaterials at the molecular level. These simulations enable researchers to investigate the interactions of biological molecules, providing insights into properties such as water absorption on biopolymer surfaces and their interactions with solid surfaces. This understanding is crucial for assessing biomaterials used in personalized therapies[5].
Additionally, agent-based modeling (ABM) offers a robust framework for simulating the interactions of biological entities, especially in oncology. ABM allows for the modeling of complex behaviors and emergent phenomena in biological systems, which can lead to improved diagnostic and therapeutic strategies. The literature review by Stephan et al. (2024) emphasizes the potential of ABM to enhance our understanding of cancer dynamics and treatment responses[6].
The integration of computational modeling in biomedical research not only enhances our understanding of biological processes but also paves the way for advancements in personalized medicine. By utilizing various modeling techniques, researchers can create more effective and tailored therapeutic approaches, ultimately leading to improved health outcomes and a more humane research paradigm that aligns with ethical standards[1][5][13].
4 Methodologies in Computational Modeling
4.1 Statistical Mechanics and Thermodynamics
Computational modeling plays a crucial role in simulating biological processes, employing various methodologies to provide insights into the complexities of biological systems. These methodologies often incorporate principles from statistical mechanics and thermodynamics to enhance the understanding of biological phenomena at different scales.
One primary approach is the development of computational models that simulate hemostasis and thrombosis, providing quantitative characterizations of thrombus development. These models utilize simulations of coagulation reactions, platelet activation, and fibrinogen assembly, which have shown close agreement with experimental data. By integrating various processes involved in hemostasis and thrombosis, these models can simulate thrombus development in both temporal and spatial dimensions, offering insights that qualitative biological models may not reveal (Xu et al., 2011) [18].
In the realm of cellular processes, molecular simulations are employed to mimic conditions within different cellular compartments. Various groups have developed molecular models that balance performance and accuracy, often simplifying atomistic degrees of freedom to explore slow processes effectively. These simulations have provided valuable insights into macromolecule diffusion, nuclear body formation, and the dynamics of genetic material, while also addressing challenges related to simulating complex biological processes over extended timescales (Trovato & Fumagalli, 2017) [8].
Advancements in computational power have further facilitated the simulation of protein dynamics, which are essential for understanding biological mechanisms. Recent molecular simulations have yielded models that align increasingly with experimental observations, enabling researchers to formulate new hypotheses and design further experiments. These simulations are particularly beneficial in elucidating the genetic, thermodynamic, and functional behaviors of biological processes, highlighting the synergy between computational methods and experimental data (Dodson et al., 2008) [19].
Moreover, the integration of multiscale computational models has proven essential in capturing the intricate interactions across different biological scales. For instance, in the study of bone remodeling, models exist at organ, tissue, and cellular levels, each addressing specific aspects of bone dynamics. However, these models often operate in isolation, which limits their interpretative power. A proposed computational framework aims to integrate these various scales, enhancing the understanding of bone metabolism and the dynamic behavior of bone under different stimuli (Webster & Müller, 2011) [20].
In conclusion, computational modeling employs methodologies rooted in statistical mechanics and thermodynamics to simulate biological processes. By bridging experimental observations and theoretical frameworks, these models enhance our understanding of complex biological systems, leading to novel insights and experimental strategies that can significantly impact the field of biology and medicine.
4.2 Machine Learning Applications
Computational modeling serves as a crucial tool for simulating biological processes, enabling researchers to understand complex interactions within biological systems. Various methodologies have been developed to enhance the accuracy and efficiency of these simulations, particularly through the integration of machine learning (ML) techniques.
Agent-based modeling (ABM) is one prominent approach highlighted in recent literature, particularly in oncology. This methodology allows for the simulation of individual biological entities and their interactions, providing insights into the emergent behaviors of complex systems. The use of ABM facilitates the exploration of potential interactions between biological elements, offering a promising avenue for understanding intricate biological processes and predicting their behavior under diverse conditions (Stephan et al. 2024) [6].
In addition to ABM, surrogate machine learning models have gained traction as a means to bridge the gap between mechanistic biological models and computational efficiency. Mechanistic models, while comprehensive, often require significant computational resources, which can limit their applicability for real-time simulations. Surrogate ML models, on the other hand, can approximate the behavior of these complex models while drastically reducing computational demands. This capability makes it feasible to simulate biological processes on standard computing platforms, thereby expanding the accessibility of sophisticated modeling techniques to a broader range of researchers (Gherman et al. 2023) [21].
The integration of computational models across multiple biological scales is another critical aspect of simulating biological processes. For instance, multiscale computational models have been employed to identify gaps in our understanding of biological systems and optimize cellular processes. These models are designed to mimic the flow of biological information from the genome to the phenome, which is essential for predicting how organisms will respond in untested environments. The challenge lies in effectively connecting models across various biological, temporal, and computational scales to generate comprehensive insights (Benes et al. 2020) [22].
Furthermore, computational modeling has advanced to include the simulation of intracellular processes, utilizing object-oriented approaches to construct abstractions of biological objects. This allows for the observation of emergent properties as these objects interact within a simulated environment, thus providing valuable insights into the complexity of biological systems (Johnson et al. 2004) [7].
In summary, computational modeling employs a variety of methodologies, including agent-based modeling and surrogate machine learning, to simulate biological processes effectively. These approaches not only enhance our understanding of complex biological interactions but also enable the exploration of hypotheses that may be challenging to investigate experimentally. As computational tools continue to evolve, their integration with biological research promises to yield significant advancements in the field of systems biology.
4.3 Agent-Based Modeling Techniques
Computational modeling serves as a crucial methodology for simulating biological processes, particularly through the use of agent-based modeling (ABM) techniques. ABMs offer a unique framework that captures the complexity and dynamism inherent in biological systems by modeling individual entities, referred to as agents, which interact based on defined rules. This bottom-up approach allows for the emergent behavior of the system to be studied as a whole, reflecting the interactions and heterogeneous nature of biological components.
One of the primary advantages of ABMs is their ability to accommodate the continuous features of biological environments while recognizing the flexibility and diversity of component interactions. Traditional mathematical models often assume homogeneity among components, which can lead to inaccuracies in systems characterized by dynamic interactions among heterogeneous agents. ABMs address this limitation by allowing for a more realistic representation of biological systems, where individual agents can exhibit diverse behaviors and properties [23].
ABMs have been particularly effective in translational systems biology, where they help translate mechanistic knowledge from basic science into computational models that can represent disease processes. This is especially pertinent in the study of inflammation, cancer, and other complex biological phenomena. The rule-based and spatially explicit nature of ABMs enables researchers to model the interactions among individual cells and their environments, thus capturing the multi-scale dynamics of cellular behavior and disease progression [24].
Recent advancements in computational power and modeling platforms have further enhanced the utility of ABMs. For instance, the development of high-performance simulation engines, such as BioDynaMo, has significantly improved the speed and scalability of ABM simulations, enabling the modeling of systems with billions of agents. This advancement facilitates the exploration of larger and more complex biological systems that were previously computationally prohibitive [25].
Moreover, ABMs have shown promise in oncology, where they can simulate tumor growth and the interactions between cancer cells and their microenvironment. This modeling approach allows for the incorporation of various biological factors, such as angiogenesis, chemotherapy responses, and cellular diversity, providing insights into tumor dynamics and potential therapeutic strategies [26]. However, challenges remain in validating these models across different scales and ensuring that they accurately reflect biological realities [27].
In summary, computational modeling through agent-based techniques plays a pivotal role in simulating biological processes by providing a framework that captures the complexity and dynamism of biological interactions. The ability to model individual behaviors and interactions within a system allows researchers to gain deeper insights into the mechanisms underlying various biological phenomena, thereby advancing our understanding of health and disease.
5 Challenges and Limitations
5.1 Data Availability and Quality
Computational modeling has emerged as a pivotal tool in simulating biological processes, offering a framework to analyze and predict the behavior of complex biological systems. However, this approach is not without its challenges and limitations, particularly concerning data availability and quality.
Computational models simulate biological processes by utilizing various techniques, including molecular simulations, pharmacokinetic/pharmacodynamic frameworks, and organ-on-chip technologies. These models aim to replicate human physiological and pathological processes with greater precision than traditional animal models, which often face ethical concerns, high costs, and poor translational relevance to human biology (Mittal et al. 2025). By bridging critical gaps in predictive accuracy and enhancing opportunities for personalized medicine, computational models support drug development pipelines and help reduce late-stage failures in clinical trials (Mittal et al. 2025).
Despite their advantages, computational modeling encounters significant challenges related to data availability and quality. One of the primary issues is the inherent complexity of biological systems, which are composed of highly interconnected subunits. This complexity makes it difficult to monitor, control, and optimize bioprocesses effectively (Noll & Henkel 2020). The accuracy of computational models is heavily dependent on the quality and comprehensiveness of the input data. Inadequate or poorly curated data can lead to inaccurate simulations and unreliable predictions.
Moreover, the force-field problem, sampling problem, ensemble problem, and experimental problem are critical limitations in biomolecular modeling (van Gunsteren et al. 2006). These issues arise from the difficulty in accurately representing the interactions and dynamics of biomolecules, which can vary significantly across different biological contexts. The lack of high-quality experimental data to validate computational models further exacerbates these challenges, as many properties of biomolecular systems remain difficult to measure experimentally (Harris & Kendon 2010).
Additionally, computational simulations often rely on approximations that may not fully capture the entropic contributions to the free energy of biomolecules, limiting their ability to predict complex behaviors accurately (Harris & Kendon 2010). As such, the generation of robust and reproducible simulation outcomes necessitates the integration of experimental and computational methods, which can be hindered by differences in vocabulary and approaches between experimental physiologists and computational modelers (Gardiner et al. 2020).
In conclusion, while computational modeling serves as a transformative approach in simulating biological processes, the challenges associated with data availability and quality remain significant. Addressing these challenges through improved data curation, interdisciplinary collaboration, and the development of standardized practices will be essential for advancing the field and enhancing the reliability of computational models in biomedical research.
5.2 Model Validation and Reproducibility
Computational modeling plays a pivotal role in simulating biological processes by employing various approaches that range from molecular simulations to system-level models. These models can mimic cellular behaviors, predict drug interactions, and provide insights into complex biological phenomena. However, the field faces significant challenges, particularly in terms of model validation and reproducibility.
One primary challenge in computational modeling is the complexity of biological systems. Biological processes often involve intricate interactions across multiple scales, from molecular to cellular to organismal levels. As such, models that simplify these processes can sacrifice important details, leading to a loss of structural complexity and chemical specificity. For instance, many molecular simulations have eliminated certain degrees of freedom to explore slow processes, which can compromise the accuracy of the predictions made by these models (Trovato & Fumagalli, 2017) [8].
The validation of computational models is another critical area of concern. Establishing the credibility of these models requires a rigorous validation process that quantifies uncertainty and assesses the models' predictive capabilities. A framework proposed by Patterson and Whelan (2017) categorizes models based on their testability and epistemic foundation, suggesting that different validation approaches should be employed depending on whether a model is deemed testable or untestable [28]. This complexity is compounded by the inherent variability of biological systems, making it difficult to ascertain the reliability of model outputs.
Reproducibility is closely linked to validation and is a significant challenge in computational biology. The proliferation of computational models has highlighted the need for improved accessibility and interoperability of these models. Niarakis et al. (2022) emphasize that effective model annotation in standardized formats is essential for enhancing the reusability of computational models in systems biology, thus aiding in the reproducibility of virtual experiments [29]. The lack of widely accepted criteria for model validation in systems biology further complicates this issue, as it hampers the ability to compare and reproduce findings across different studies (Gross & MacLeod, 2017) [30].
Moreover, as computational modeling becomes increasingly integrated into biomedical research, there is a pressing need for interdisciplinary collaboration between experimentalists and computational scientists. This collaboration is essential to bridge the gap between theoretical models and empirical data, fostering a more iterative process of model development and validation (Gardiner et al., 2020) [31].
In conclusion, while computational modeling holds great promise for simulating biological processes, the challenges of model validation and reproducibility remain significant hurdles. Addressing these issues requires ongoing efforts to standardize validation practices, enhance model accessibility, and promote collaboration across disciplines, ensuring that computational models can reliably contribute to our understanding of complex biological systems.
5.3 Computational Resource Constraints
Computational modeling has emerged as a pivotal tool for simulating biological processes, particularly in the context of understanding complex systems at molecular and cellular levels. However, this approach is not without its challenges and limitations, particularly concerning computational resource constraints.
One significant challenge in computational modeling is the inherent complexity of biological systems. Biological processes are often characterized by a multitude of interacting components, such as proteins, nucleic acids, and cellular structures, which necessitate detailed and accurate models. The configurational complexity of biomolecules, for instance, implies that entropic contributions to free energy play a critical role in their behavior. This complexity requires simulations that account for all interatomic interactions, which can be computationally intensive [32].
Moreover, traditional computational methods often struggle with the limitations imposed by classical computing architectures. The speed and efficiency of digital silicon computers have reached a plateau, hindering progress in simulating intricate biological phenomena. As a result, researchers are exploring alternative computational paradigms, such as quantum computing, which promises to significantly enhance the speed and efficiency of simulations by allowing parallel processing of numerous computational paths [33].
Another critical aspect of computational resource constraints is the issue of sampling in molecular simulations. The rugged energy landscape of complex biomolecules can lead to limited conformational sampling, making it difficult to capture the full range of biologically relevant states. Enhanced sampling techniques have been developed to address this issue, but they often require substantial computational resources and can still fall short of adequately exploring the potential energy surface [34].
Furthermore, the scalability of computational models poses another layer of difficulty. As biological systems are modeled at larger scales—incorporating more cells or simulating longer timeframes—the computational demands increase exponentially. For instance, simulating multicellular systems or complex interactions within a human brain model is currently beyond the reach of available computational resources and may remain so for the foreseeable future [35].
In summary, while computational modeling provides invaluable insights into biological processes, it faces significant challenges related to computational resource constraints. These include the complexity of biological systems, limitations of current computing technologies, issues with sampling in molecular dynamics, and the scalability of models to represent larger biological phenomena. Addressing these challenges is essential for advancing the field of computational biology and improving our understanding of life at the molecular level.
6 Future Directions in Computational Modeling
6.1 Integrating Multi-Omics Data
Computational modeling plays a crucial role in simulating biological processes by leveraging mathematical frameworks to represent the complex interactions within biological systems. This approach enables researchers to integrate various types of biological data, particularly from multi-omics sources, which include genomics, transcriptomics, proteomics, and metabolomics. The integration of these diverse datasets is essential for achieving a comprehensive understanding of biological phenomena.
Computational models, such as genome-scale models (GEMs), convert biological reactions related to metabolism, transcription, and translation into mathematical formulations that can be optimized. This modeling process facilitates the interpretation of multi-omic data and provides insights into the systemic interactions that govern cellular functions. As noted by Dahal et al. (2020), the synthesis of knowledge through the integration of omic data with GEMs is expected to yield significant advancements in fields such as human health and metabolic engineering [36].
Moreover, the advancements in computational methodologies, including machine learning techniques like deep learning and generative adversarial networks, enhance the ability to analyze and interpret multi-omics data effectively. Luo et al. (2024) emphasize that these computational tools are vital for uncovering the intricate interactions and regulatory mechanisms underlying biological processes [37]. The integration of these methods not only addresses the challenges posed by data heterogeneity and scalability but also supports the development of robust models that can capture the complexity of biological systems.
Future directions in computational modeling are likely to focus on the continued integration of multi-omics data to create more holistic models that can simulate biological processes at various scales. For instance, the application of single-cell omics data is being explored to build high-resolution computational models that can simulate biological systems, as highlighted by Manchel et al. (2024). This approach allows for the assembly of organ-specific models, which can be further integrated to develop multi-organ systems pathophysiological models [38].
Additionally, the incorporation of artificial intelligence in handling missing data within multi-omics integration presents a promising avenue for enhancing model accuracy and robustness. Flores et al. (2023) discuss recent advances in methodologies that can effectively manage incomplete datasets, which is a common challenge in multi-omics studies [39]. These developments are crucial for advancing the predictive capabilities of computational models and ensuring that they can be applied in clinical settings for precision medicine.
In conclusion, computational modeling serves as a powerful tool for simulating biological processes by integrating multi-omics data, which enhances our understanding of complex biological interactions. The ongoing evolution of computational methodologies and the incorporation of advanced technologies will continue to shape the future of systems biology, facilitating the development of more comprehensive and predictive models that can inform research and clinical applications.
6.2 Advancements in Computational Power
Computational modeling plays a crucial role in simulating biological processes, providing insights into complex biological systems through the use of mathematical and computational frameworks. This approach allows researchers to understand, predict, and manipulate biological phenomena by simulating interactions at various biological scales, from molecular to cellular and tissue levels.
One significant advancement in computational modeling is the integration of artificial intelligence (AI) and agent-based modeling (ABM). These methodologies have been particularly beneficial in oncology, as they facilitate the understanding of cancer behaviors and improve diagnostic capabilities. Recent literature highlights the use of metaheuristic algorithms as prevalent models in this domain, which assist in addressing challenges such as knowledge structuring about biological systems, developing holistic models that capture emergent behaviors, and ensuring model validation with experimental data [6].
Furthermore, computational models serve as essential tools for interpreting data generated from high-throughput genomics and proteomics. They allow for rapid access to and sharing of knowledge through data mining, thus generating hypotheses and suggesting experimental directions [40]. The advancements in computational power have enabled the simulation of intricate biological processes, leading to a deeper understanding of cellular mechanisms and the interactions of various biological elements [41].
The future directions in computational modeling are promising, particularly with the emergence of multiscale modeling techniques that bridge biological research, data science, and clinical practice. These models can integrate data across different scales and levels of biological organization, providing a more comprehensive understanding of complex systems. As computational capabilities continue to evolve, the potential for personalized medicine and adaptive therapies will be significantly enhanced, as predictive modeling approaches become integral to therapeutic development [42].
Moreover, the application of biomolecular simulations allows researchers to gain insights into molecular-level biological processes over extended periods, thus revealing connections between molecular structures and biological functions. This is critical for advancing our understanding of various biological systems, including disease mechanisms and potential therapeutic interventions [41].
In conclusion, computational modeling is transforming the landscape of biological research by enabling detailed simulations of complex processes, facilitating the integration of data across scales, and paving the way for innovative solutions in medicine and biology. The ongoing advancements in computational power and modeling techniques are expected to drive further breakthroughs in our understanding of biological systems and their applications in health and disease.
6.3 Enhancing Model Interoperability
Computational modeling serves as a vital tool in simulating biological processes, enabling researchers to study complex biological systems and predict their behaviors under various conditions. This approach is particularly relevant in fields such as oncology, where agent-based modeling (ABM) and other computational techniques are utilized to understand interactions between biological elements and improve diagnostic capabilities (Stephan et al. 2024) [6]. The methodology encompasses a range of simulation techniques, including molecular simulations that replicate cellular environments, thereby allowing for insights into macromolecule diffusion, nuclear body formation, and genetic material processes (Trovato and Fumagalli 2017) [8].
One significant challenge in computational modeling is enhancing model interoperability. This refers to the ability of different computational models to work together seamlessly, sharing data and processes across various scales of biological organization. For instance, the integration of models that simulate processes at different temporal and spatial scales is crucial for capturing emergent properties of biological systems (Benes et al. 2020) [22]. Effective interoperability can facilitate a more holistic understanding of biological phenomena, leading to improved experimental strategies and outcomes.
To address the barriers in model interoperability, recent efforts have focused on developing standardized frameworks that ensure models are comprehensively annotated and accessible (Niarakis et al. 2022) [29]. Such initiatives aim to enhance the reusability and reproducibility of computational models, which are essential for validating findings and advancing biological research. Furthermore, the democratization of simulation technologies can encourage broader participation from biologists, enabling the collective advancement of modeling approaches that contribute to our understanding of complex biological systems (Iwasa et al. 2022) [43].
The future of computational modeling in biology lies in the integration of advanced computational techniques and the development of robust workflows that support modularity, scalability, and interoperability (Yang et al. 2019) [44]. As the field continues to evolve, the emphasis on collaborative efforts and shared knowledge will be pivotal in overcoming existing challenges and unlocking the full potential of computational modeling in simulating biological processes. This evolution is expected to lead to significant advancements in areas such as drug development, personalized medicine, and the understanding of intricate biological interactions.
7 Conclusion
Computational modeling has emerged as a transformative tool in the field of biomedical research, enabling researchers to simulate and analyze complex biological processes with remarkable precision. The findings from this report highlight several key aspects of computational modeling, including its diverse methodologies, applications in disease mechanism simulation, drug discovery, and personalized medicine approaches. Notably, techniques such as agent-based modeling, molecular dynamics simulations, and multiscale modeling have significantly enhanced our understanding of intricate biological interactions and therapeutic strategies. Despite the remarkable advancements, challenges related to data availability, model validation, and computational resource constraints persist. The future of computational modeling lies in the integration of multi-omics data, advancements in computational power, and enhanced model interoperability. As these challenges are addressed, computational modeling is poised to play an even more critical role in unraveling the complexities of biological systems and improving patient outcomes in healthcare. Continued interdisciplinary collaboration and innovation in computational methodologies will be essential to fully realize the potential of this dynamic field.
References
- [1] Rahul Mittal;Alan Ho;Harini Adivikolanu;Muskaan Sawhney;Joana R N Lemos;Mannat Mittal;Khemraj Hirani. Exploring the potential of computer simulation models in drug testing and biomedical research: a systematic review.. Frontiers in pharmacology(IF=4.8). 2025. PMID:41069606. DOI: 10.3389/fphar.2025.1644907.
- [2] Seth Michelson;Mike Cole. The future of predictive biosimulation in drug discovery.. Expert opinion on drug discovery(IF=4.9). 2007. PMID:23484759. DOI: 10.1517/17460441.2.4.515.
- [3] Neil Kumar;Bart S Hendriks;Kevin A Janes;David de Graaf;Douglas A Lauffenburger. Applying computational modeling to drug discovery and development.. Drug discovery today(IF=7.5). 2006. PMID:16935748. DOI: 10.1016/j.drudis.2006.07.010.
- [4] David Gavaghan;Alan Garny;Philip K Maini;Peter Kohl. Mathematical models in physiology.. Philosophical transactions. Series A, Mathematical, physical, and engineering sciences(IF=3.7). 2006. PMID:16608698. DOI: 10.1098/rsta.2006.1757.
- [5] Ramón Garduño-Juárez;David O Tovar-Anaya;Jose Manuel Perez-Aguilar;Luis Fernando Lozano-Aguirre Beltran;Rafael A Zubillaga;Marco Antonio Alvarez-Perez;Eduardo Villarreal-Ramirez. Molecular Dynamic Simulations for Biopolymers with Biomedical Applications.. Polymers(IF=4.9). 2024. PMID:39000719. DOI: 10.3390/polym16131864.
- [6] Simon Stephan;Stéphane Galland;Ouassila Labbani Narsis;Kenji Shoji;Sébastien Vachenc;Stéphane Gerart;Christophe Nicolle. Agent-based approaches for biological modeling in oncology: A literature review.. Artificial intelligence in medicine(IF=6.2). 2024. PMID:38703466. DOI: 10.1016/j.artmed.2024.102884.
- [7] Colin G Johnson;Jacki P Goldman;William J Gullick. Simulating complex intracellular processes using object-oriented computational modelling.. Progress in biophysics and molecular biology(IF=4.5). 2004. PMID:15302205. DOI: 10.1016/j.pbiomolbio.2003.11.001.
- [8] Fabio Trovato;Giordano Fumagalli. Molecular simulations of cellular processes.. Biophysical reviews(IF=3.7). 2017. PMID:29185136. DOI: 10.1007/s12551-017-0363-6.
- [9] Nikolay V Dokholyan. Studies of folding and misfolding using simplified models.. Current opinion in structural biology(IF=7.0). 2006. PMID:16413773. DOI: 10.1016/j.sbi.2006.01.001.
- [10] Martin Meier-Schellersheim;Iain D C Fraser;Frederick Klauschen. Multiscale modeling for biologists.. Wiley interdisciplinary reviews. Systems biology and medicine(IF=7.9). 2009. PMID:20448808. DOI: 10.1002/wsbm.33.
- [11] Kathryn Atwell;Sara-Jane Dunn;James M Osborne;Hillel Kugler;E Jane Albert Hubbard. How computational models contribute to our understanding of the germ line.. Molecular reproduction and development(IF=3.0). 2016. PMID:27627621. DOI: 10.1002/mrd.22735.
- [12] Esra Neufeld;Dominik Szczerba;Nicolas Chavannes;Niels Kuster. A novel medical image data-based multi-physics simulation platform for computational life sciences.. Interface focus(IF=4.0). 2013. PMID:24427518. DOI: 10.1098/rsfs.2012.0058.
- [13] Gary An;John Bartels;Yoram Vodovotz. In Silico Augmentation of the Drug Development Pipeline: Examples from the study of Acute Inflammation.. Drug development research(IF=4.2). 2011. PMID:21552346. DOI: 10.1002/ddr.20415.
- [14] Marzio Pennisi;Giulia Russo;Valentina Di Salvatore;Saverio Candido;Massimo Libra;Francesco Pappalardo. Computational modeling in melanoma for novel drug discovery.. Expert opinion on drug discovery(IF=4.9). 2016. PMID:27046143. DOI: 10.1080/17460441.2016.1174688.
- [15] Si-Sheng Ou-Yang;Jun-Yan Lu;Xiang-Qian Kong;Zhong-Jie Liang;Cheng Luo;Hualiang Jiang. Computational drug discovery.. Acta pharmacologica Sinica(IF=8.4). 2012. PMID:22922346. DOI: 10.1038/aps.2012.109.
- [16] Tope T Odunitan;Oluwatosin A Saibu;Boluwatife T Apanisile;Damilola A Omoboyowa;Toheeb A Balogun;Adeyoola V Awe;Temitope M Ajayi;Grace V Olagunju;Fatimah M Mahmoud;Modinat Akinboade;Catherine B Adeniji;Waliu O Abdulazeez. Integrating biocomputational techniques for Breast cancer drug discovery via the HER-2, BCRA, VEGF and ER protein targets.. Computers in biology and medicine(IF=6.3). 2024. PMID:38000249. DOI: 10.1016/j.compbiomed.2023.107737.
- [17] Lixia Yao;James A Evans;Andrey Rzhetsky. Novel opportunities for computational biology and sociology in drug discovery.. Trends in biotechnology(IF=14.9). 2009. PMID:19674801. DOI: 10.1016/j.tibtech.2009.06.003.
- [18] Zhiliang Xu;Malgorzata Kamocka;Mark Alber;Elliot D Rosen. Computational approaches to studying thrombus development.. Arteriosclerosis, thrombosis, and vascular biology(IF=7.4). 2011. PMID:21325672. DOI: 10.1161/ATVBAHA.110.213397.
- [19] Guy G Dodson;David P Lane;Chandra S Verma. Molecular simulations of protein dynamics: new windows on mechanisms in biology.. EMBO reports(IF=6.2). 2008. PMID:18246106. DOI: 10.1038/sj.embor.7401160.
- [20] Duncan Webster;Ralph Müller. In silico models of bone remodeling from macro to nano--from organ to cell.. Wiley interdisciplinary reviews. Systems biology and medicine(IF=7.9). 2011. PMID:20740496. DOI: 10.1002/wsbm.115.
- [21] Ioana M Gherman;Zahraa S Abdallah;Wei Pang;Thomas E Gorochowski;Claire S Grierson;Lucia Marucci. Bridging the gap between mechanistic biological models and machine learning surrogates.. PLoS computational biology(IF=3.6). 2023. PMID:37079494. DOI: 10.1371/journal.pcbi.1010988.
- [22] Bedrich Benes;Kaiyu Guan;Meagan Lang;Stephen P Long;Jonathan P Lynch;Amy Marshall-Colón;Bin Peng;James Schnable;Lee J Sweetlove;Matthew J Turk. Multiscale computational models can guide experimentation and targeted measurements for crop improvement.. The Plant journal : for cell and molecular biology(IF=5.7). 2020. PMID:32053236. DOI: 10.1111/tpj.14722.
- [23] Himanshu Kaul;Yiannis Ventikos. Investigating biocomplexity through the agent-based paradigm.. Briefings in bioinformatics(IF=7.7). 2015. PMID:24227161. DOI: 10.1093/bib/bbt077.
- [24] Yoram Vodovotz;Gary An. Agent-based models of inflammation in translational systems biology: A decade later.. Wiley interdisciplinary reviews. Systems biology and medicine(IF=7.9). 2019. PMID:31260168. DOI: 10.1002/wsbm.1460.
- [25] Lukas Breitwieser;Ahmad Hesam;Jean de Montigny;Vasileios Vavourakis;Alexandros Iosif;Jack Jennings;Marcus Kaiser;Marco Manca;Alberto Di Meglio;Zaid Al-Ars;Fons Rademakers;Onur Mutlu;Roman Bauer. BioDynaMo: a modular platform for high-performance agent-based simulation.. Bioinformatics (Oxford, England)(IF=5.4). 2022. PMID:34529036. DOI: 10.1093/bioinformatics/btab649.
- [26] Zhihui Wang;Joseph D Butner;Romica Kerketta;Vittorio Cristini;Thomas S Deisboeck. Simulating cancer growth with multiscale agent-based modeling.. Seminars in cancer biology(IF=15.7). 2015. PMID:24793698. DOI: .
- [27] Nicolò Cogno;Cristian Axenie;Roman Bauer;Vasileios Vavourakis. Agent-based modeling in cancer biomedicine: applications and tools for calibration and validation.. Cancer biology & therapy(IF=4.6). 2024. PMID:38678381. DOI: 10.1080/15384047.2024.2344600.
- [28] Eann A Patterson;Maurice P Whelan. A framework to establish credibility of computational models in biology.. Progress in biophysics and molecular biology(IF=4.5). 2017. PMID:27702656. DOI: 10.1016/j.pbiomolbio.2016.08.007.
- [29] Anna Niarakis;Dagmar Waltemath;James Glazier;Falk Schreiber;Sarah M Keating;David Nickerson;Claudine Chaouiya;Anne Siegel;Vincent Noël;Henning Hermjakob;Tomáš Helikar;Sylvain Soliman;Laurence Calzone. Addressing barriers in comprehensiveness, accessibility, reusability, interoperability and reproducibility of computational models in systems biology.. Briefings in bioinformatics(IF=7.7). 2022. PMID:35671510. DOI: 10.1093/bib/bbac212.
- [30] Fridolin Gross;Miles MacLeod. Prospects and problems for standardizing model validation in systems biology.. Progress in biophysics and molecular biology(IF=4.5). 2017. PMID:28089814. DOI: 10.1016/j.pbiomolbio.2017.01.003.
- [31] Bruce S Gardiner;David W Smith;Chang-Joon Lee;Jennifer P Ngo;Roger G Evans. Renal oxygenation: From data to insight.. Acta physiologica (Oxford, England)(IF=5.6). 2020. PMID:32012449. DOI: 10.1111/apha.13450.
- [32] Sarah A Harris;Vivien M Kendon. Quantum-assisted biomolecular modelling.. Philosophical transactions. Series A, Mathematical, physical, and engineering sciences(IF=3.7). 2010. PMID:20603369. DOI: 10.1098/rsta.2010.0087.
- [33] Viv Kendon. Quantum computing using continuous-time evolution.. Interface focus(IF=4.0). 2020. PMID:33178417. DOI: 10.1098/rsfs.2019.0143.
- [34] Hongxing Lei;Yong Duan. Improved sampling methods for molecular simulation.. Current opinion in structural biology(IF=7.0). 2007. PMID:17382533. DOI: 10.1016/j.sbi.2007.03.003.
- [35] Roland R Netz;William A Eaton. Estimating computational limits on theoretical descriptions of biological cells.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 2021. PMID:33495364. DOI: 10.1073/pnas.2022753118.
- [36] Sanjeev Dahal;James T Yurkovich;Hao Xu;Bernhard O Palsson;Laurence Yang. Synthesizing Systems Biology Knowledge from Omics Using Genome-Scale Models.. Proteomics(IF=3.9). 2020. PMID:32579720. DOI: 10.1002/pmic.201900282.
- [37] Yunqing Luo;Chengjun Zhao;Fei Chen. Multiomics Research: Principles and Challenges in Integrated Analysis.. Biodesign research(IF=4.7). 2024. PMID:39990095. DOI: 10.34133/bdr.0059.
- [38] Alexandra Manchel;Michelle Gee;Rajanikanth Vadigepalli. From sampling to simulating: Single-cell multiomics in systems pathophysiological modeling.. iScience(IF=4.1). 2024. PMID:39628578. DOI: 10.1016/j.isci.2024.111322.
- [39] Javier E Flores;Daniel M Claborne;Zachary D Weller;Bobbie-Jo M Webb-Robertson;Katrina M Waters;Lisa M Bramer. Missing data in multi-omics integration: Recent advances through artificial intelligence.. Frontiers in artificial intelligence(IF=4.7). 2023. PMID:36844425. DOI: 10.3389/frai.2023.1098308.
- [40] Santo Motta;Francesco Pappalardo. Mathematical modeling of biological systems.. Briefings in bioinformatics(IF=7.7). 2013. PMID:23063928. DOI: 10.1093/bib/bbs061.
- [41] Ekambaram Gayathiri;Palanisamy Prakash;Priya Kumaravel;Jayanthi Jayaprakash;Manikkavalli Gurunathan Ragunathan;Sharmila Sankar;Saravanan Pandiaraj;Natesan Thirumalaivasan;Muthu Thiruvengadam;Rajakumar Govindasamy. Computational approaches for modeling and structural design of biological systems: A comprehensive review.. Progress in biophysics and molecular biology(IF=4.5). 2023. PMID:37821048. DOI: 10.1016/j.pbiomolbio.2023.08.002.
- [42] Christian Baumgartner. Computational modeling and simulation in oncology.. Clinical and translational medicine(IF=6.8). 2025. PMID:40910400. DOI: 10.1002/ctm2.70456.
- [43] Janet H Iwasa;Blair Lyons;Graham T Johnson. The dawn of interoperating spatial models in cell biology.. Current opinion in biotechnology(IF=7.0). 2022. PMID:36402095. DOI: 10.1016/j.copbio.2022.102838.
- [44] Pei-Chi Yang;Shweta Purawat;Pek U Ieong;Mao-Tsuen Jeng;Kevin R DeMarco;Igor Vorobyov;Andrew D McCulloch;Ilkay Altintas;Rommie E Amaro;Colleen E Clancy. A demonstration of modularity, reuse, reproducibility, portability and scalability for modeling and simulation of cardiac electrophysiology using Kepler Workflows.. PLoS computational biology(IF=3.6). 2019. PMID:30849072. DOI: 10.1371/journal.pcbi.1006856.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Computational Modeling · Biological Processes · Drug Discovery · Personalized Medicine · Disease Mechanisms
© 2025 MaltSci
