Developing a Framework for Multi-Scale Modeling of the Digital
Patient: Insights from Current Status and Future Directions
C. Donald Combs
*
, Lubna Pinky, Chathurani Ranathunge, Sagar S. Patel, Taryn Cuper,
Robert K. Armstrong and Robert J. Alpino
School of Health Professions, Eastern Virginia Medical School, Norfolk, VA 23507, U.S.A.
Keywords: Digital Patient, Systems Biology, Physics-Based Modeling, Artificial Intelligence, Bigdata, Bio-Informatics,
Pharmaceutical Research, Virtual Clinical Trials, Personalized Medicine.
Abstract: The Digital Patient is an analytic platform that has the potential to transform personal and public healthcare,
pharmaceutical research, medical device development, and patient and professional education. It is the
ultimate big data project in healthcare; however, its power will derive not from the volume of data, but from
the successful and efficient integration of disparate sources of data into a validated and reliable computational
model of combined biological processes, social context and treatment efficacy. That integration, successively,
is largely dependent on the evolving theoretical approaches known as systems biology and physics-based
modeling that lead to the successful meshing of multi-scale models.
1 WHAT IS A DIGITAL PATIENT?
The Digital Patient is a comprehensive approach to
providing a computational platform for personalized
medicine. It is a digital representation of a person’s
‘health’ and ‘disease’ status or in another word, a
whole-body system. It may include computer models
of the mechanical, physical, and biochemical
functions of a living human body calibrated by multi-
scale and length data collected from the multiple
physiological levels. This is not only the ultimate big
data project, but also the ultimate technical challenge
in medicine. When executed, it can be used as a
powerful in silico (in silico means carried out in the
computer, which is in contrast to in vitro (on the
bench), ex vivo (outside the living organism), or in
vivo (inside the living organism)) decision support
technology that can be customized to represent each
one of us, individually and/or collectively (C. D.
Combs et al., 2015; Dıaz-Zuccarini et al., 2015;
Parodi, 2015). It is an integrated approach for
achieving a broader, more systematic understanding
of the human body in a single computational platform.
One of the key aspects to the project is the need
for a new analytic framework for understanding the
whole-body, that is, being able to deal with the
complexity of physiology by creating and integrating
*
Corresponding author
properly annotated, curated, validated and
documented modules of individual organ systems.
Ultimately, more complex models are expected to
merge upon linking these modules semantically to
represent the whole-body system. In the construction
of a Digital Patient framework, significantly
important things to consider are what level of detail is
necessary, and more importantly, how to accurately
predict the efficacy of a patient-specific treatment. In
addition, the processes for achieving a Digital Patient
framework involve generation of data and
information, biomedical information management,
knowledge-driven and data-driven computational
modeling (i.e., in silico models based on prior
knowledge on cause-and-effect relationships, and/or
nature of data), clinical user interface, and end
translation and adoption.
1.1 Integrative Whole-Body View
The human body is the ultimate efficient machine
comprised of multiple organs, tissues, and cellular
complexes that interact to maintain a homeostasis.
Pathology is due to alterations in one or more tissues
or systems or in a single biological process (Talbot et
al., 2016). Alterations in one system often induce
changes in the physiology of other systems.
Combs, C., Pinky, L., Ranathunge, C., Patel, S., Cuper, T., Armstrong, R. and Alpino, R.
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions.
DOI: 10.5220/0012140700003546
In Proceedings of the 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications (SIMULTECH 2023), pages 143-158
ISBN: 978-989-758-668-2; ISSN: 2184-2841
Copyright
c
2023 by SCITEPRESS Science and Technology Publications, Lda. Under CC license (CC BY-NC-ND 4.0)
143
Therefore, the development of an integrated model of
human physiology is essential for the understanding
of how molecular, cellular, organ and system levels
interact for a total physiological response (An &
Cockrell, 2022; Hester et al., 2011). However,
biologists have traditionally sought to understand
living entities by investigating their constituent parts
in a controlled environment, i.e., in a reductionist
way, popularly known as reductionism (Morchio,
1991; Schaffner, 1976; Woese, 2004). For example,
first they isolated the individual genes, proteins, or
signaling molecules, and then they studied them
individually to learn everything they could about the
structure and function of that single biological entity,
without necessarily considering how they interact
with one another (Bertolaso, 2022; Bricmont, 2022;
Brigandt, 2013; Kuijper, 2022; Mazzocchi, 2008). In
contrast, the Digital Patient concept is based on an
integrated approach for achieving a broader, and
more systematic understanding of the human body,
and how it interacts with its environment, i.e., social
and behavioral factors (e.g., age, gender, body
weight, genetic make-up, lifestyle, etc. (C. D. Combs
& Combs, 2014).
Future healthcare will rely more and more on data
from monitoring devices and in some cases,
implanted medical devices such as wearable
biosensors. Interpretation of data, and their
therapeutic application requires knowledge that is
much more integrated and personalized than is
currently available (C. D. Combs & Combs, 2014;
Hatzikirou et al., 2012; D. P. Nickerson et al., 2015;
D. P. Nickerson et al., 2020; Tolk et al., 2015). Most
chronic diseases involve multiple organ systems.
Therefore, it is crucial to understand how the body
works as an integrative whole during homeostasis,
and at the same time to be able to utilize our detailed
knowledge of individual organs. Moreover, in order
to represent a comprehensive Digital Patient, social
systems and environmental factors must ultimately be
integrated into this analytic framework (C. D. Combs,
2017).
1.2 Biology View
Focusing more directly on converging different levels
of biological systems that are essential to the Digital
Patient framework is the discipline of systems
biology and its applications. Systems biology is an
integrative and interdisciplinary approach in contrast
to the traditional reductionist nature of biology
(Bertolaso, 2022; Bricmont, 2022; Brigandt, 2013;
Kuijper, 2022; Mazzocchi, 2008; Schaffner, 1976;
Woese, 2004). It attempts to explain complex
biological systems that include biochemical systems
(e.g., enzyme activity regulation and flux in
metabolic pathways), cellular processes (e.g., gene
regulation, protein transport, signaling pathways, the
cell cycle, and apoptosis), cell-cell interaction (e.g.,
cell–cell signaling), as well as cell differentiation and
organismal development (Boogerd et al., 2007;
Kitano, 2001; Klipp et al., 2016), using a variety of
conceptual and experimental methods such as
genomics, transcriptomics, proteomics, molecular
biology, cell biology, and carefully developed animal
models. Thus, systems biology has the potential to
provide valuable insights into the physiological
workings of the human body. The current goal of
systems biology research is to utilize scientific
advancements from the past two decades, such as
genomics and proteomics, in an effort to develop
targeted therapeutic strategies (Fitzgerald et al., 2006;
Khoo et al., 2021; Kohl et al., 2010). Over the past
two decades, sequencing technologies (e.g., Next-
Generation Sequencing, Whole-Genome Sequencing,
and etc.) have made remarkable progress (Hartman et
al., 2019). As these new technologies continue to
develop, the costs associated with sequencing have
decreased dramatically, making these technologies
more affordable and accessible. Rapid advances in
high-throughput technologies coupled with the
decrease in sequencing costs have led to generation
of massive amounts of biological data, and in turn, the
abundance of biological data has made data
integration approaches increasingly popular in the
field of systems biology
Systems biology not only addresses interactions
in biological systems at different scales of biological
organization, but also is characterized by quantitative
descriptions of biological processes, using a variety
of statistical and computational techniques (Baccam
et al., 2006; Karr et al., 2022; Perelson et al., 1996).
As was stated before, biological systems consist of a
large number of functionally diverse components,
which interact highly selectively and often
nonlinearly to produce coherent behaviors (Klipp et
al., 2005; Likić et al., 2010). These components may
be individual molecules (e.g., signaling or metabolic
networks), assemblies of interacting complexes, sets
of physical factors that guide the development of an
organism (genes, mRNA, associated proteins and
protein complexes), cells in tissues or organs, and
even entire organisms in ecological communities.
What is common to all these examples is the sheer
number of components, and their selective, non-linear
interactions that render the behaviors of these systems
beyond the intuitive grasp. Mathematical models of
biological systems are most suitable and are
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
144
increasingly being used to represent our knowledge
about these systems (Iglesias & Ingalls, 2010; Ingalls,
2013). Thus, systems biology combines the
development and application of predictive
mathematical and computational modeling with
experimental studies. The quantitative techniques,
such as high-resolution microscopy, mass
spectrometry, flow cytometry, and more, that are
employed to incorporate multiple spatial and
temporal scales are consistent with the integrative
perspective of the Digital Patient framework.
1.3 Physics-Based Multi-Scale
Modeling View
Multi-scale modeling (MSM) integrates multiple
physiological processes across different length and
time scales to provide improved predictive and
individualized healthcare. The highly complex nature
of biomedical systems resulting from several distinct
factors includes the non-linearity and redundancy of
physiological states. They arise from multiple
mechanisms simultaneously pushing and pulling on
clinically relevant and/or experimentally observable
response variables (Vieira & Laubenbacher, 2022).
The concept of non-linearity states that many high-
level and integrative behaviors of the biological
system cannot be described solely through the sum of
inputs from basic processes (Auslander et al., 1972;
Oster & Perelson, 1973). The resulting heterogeneity
along with the disparate time constraints further
stimulates individual variability leading to distinct
disease outcomes across the population. Despite the
extensive complexities of biological and biomedical
systems, researchers are using both linear and non-
linear sophisticated biological and physiological
models to better understand fundamental relationships
within the human body (Bauer et al., 2009;
Beauchemin & Handel, 2011; Hester et al., 2011).
MSM, also known as knowledge-driven
modeling, mechanistic modeling, hypothesis- based
modeling, or physics-based modeling, is an equation-
based approach based on ordinary differential
equations, partial differential equations, stochastic
processes, agent-based modeling, etc. that
incorporates nonlinear coupled processes that occur
at different temporal and/or spatial scales, and lead to
a systematic integration of knowledge at the
molecular, cellular, and tissue levels (Altan-Bonnet et
al., 2020; Coveney & Fowler, 2005; Perelson &
Weisbuch, 1997; Pinky et al., 2021; Pinky &
Dobrovolny, 2017). Since biological entities have a
complex hierarchy of structure, mechanical
properties, and behavior across spatial and temporal
scales, MSM supports this integrative view by
explicitly defining the biological hypothesis or the
primary mechanisms and formalizing it into
mathematical equations (Harline et al., 2021; Pruett
& Hester, 2015; Radhakrishnan, 2020). This
approach assumes that behavior at larger scales
emerges naturally from the processes occurring at
smaller scales; in other words, embedding processes
at a small scale into the larger scales leads to a
prediction of overall system behavior (Gold et al.,
2019; Meier-Schellersheim et al., 2009). The
relationship between the biological and mathematical
theory determines the balance of phenomenology and
quantitative prediction. This step may become
iterative as the modeler balances the complexity of
the biological inputs included in the system with the
level of mathematical theory most suitable to form a
“minimum model” Kamerlin & Warshel, 2011;
Laubenbacher et al., 2021; Radhakrishnan, 2021;
Sego et al., 2022).
Working through biological and mathematical
theory should also reveal what data can be collected
from the system to inform the model construction,
i.e., what mechanisms and relationships are known to
exist, and which will be inferred, and what outcomes
or predictions will be tested. In addition, they can take
into account the influence of behavioral and social
context on the whole biological system. Thus, the
model connects the association of genetics to
proteins, proteins to cells, cells to organs, organs to
complete whole-body systems, as well as systems to
the organism itself and to the surrounding social
environment. For example, social and behavioral
context refers to taking into account the
understanding of the impact of the behavior of family
and friends on individual lifestyle choices and health
(C. D. Combs et al., 2015; Tolk et al., 2015).
1.4 Artificial Intelligence View
Artificial Intelligence (AI) is ideally suited to
discovering meaningful patterns in big data that may
otherwise escape human attention, and can offer a
more efficient means of understanding systems
dynamics and hence structuring preventive care
strategies more efficiently. Machine learning (ML)
method, a subset of AI tools, is at its core a data-
driven process, and defined as in silico models that
develop a predictor automatically for the data without
making any causal assumptions (Alber et al., 2019;
Peng et al., 2021). Unlike ML, MSM is generally
considered to be a theory-driven process and a more
traditional approach. It starts with developing
hypotheses, followed by collecting and analyzing data
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions
145
to test these hypotheses and drawing theoretical
conclusions based on the results (Pruett & Hester,
2015). It focuses on identifying abstract constructs
and the relationships among them. However, due to
the complexity of the environments and processes that
generate data, there may not be a strong theoretical
basis for the questions being studied ( T e i c h e r t
et al.,2019). In contrast, data-driven research
involves analyzing data to extract scientifically
interesting insights (i.e., robust correlations between
sets of variables) by applying analytical techniques
and modes of reasoning based on the data available
rather than prediction based on theory. It is worth
noting that some instances of ML in the literature are
described as theory-guided and seek causality by
integrating physics-based mechanistic models at
multiple temporal and spatial scales with big data
(Giansanti, 2022). In this way, ML can make up for
any unknown physics by learning the dynamics of the
system overall and thereby possibly classify patients
into specific treatment regimens. However, this
approach benefits from the knowledge and
mechanistic insights achieved through MSM to
develop novel learning algorithms with greater
robustness, data-efficiency, and generalization of
performance in data-limited situations (Peng et al.,
2021). As such, perhaps this approach can be
described as a meeting of ML and MSM to optimize
the contributions of both techniques. There are many
examples of data-driven MSM that appear to involve
the use of ML to optimize the parameter estimation
and functions. Perhaps these cases can also be
described as precursors to the combination of MSM
and ML (Alber et al., 2019; Maass et al., 2018).
In general, MSM provides insight into biological,
biomedical, and behavioral systems at a high level of
resolution and precision, which naturally produces
massive output data sets. Because computational
physiological simulations, e.g., physics-based MSM,
are too slow for clinical application, AI tools can
provide ways to speed up Digital Patient workflows.
For example, using ML methods one can develop a
simplified surrogate model (i.e., a statistical model to
accurately approximate simulation output) to reduce
complexity (Kennedy & O’Hagan, 2001). In the
context of the Digital Patient, MSM and ML
complement one another with respect to biological,
biomedical, and behavioral research and are possibly
even more powerful when combined (Costello &
Martin, 2018; Linka et al., 2020; Muzio et al., 2021).
The most sophisticated Digital Patient models are
expected to be self-improving. These models can
continuously monitor divergence between predictions
and observations, and use these divergences to
improve their own accuracy. Their deployment would
enable rapid refinement and improvement, especially
if they were designed in a modular fashion to permit
the parallel development and optimization of their
component sub-models (Maass et al., 2018).
1.5 Personalized Medicine View
Physiology is a basic medical science as knowledge
of normal functions of the body is the basis for
understanding diseases and identifying targets for
effective treatment (Sherwood, 2015). Just as
physiology is a branch of biology, systems
physiology, systems medicine and personalized
medicine are subsets of systems biology. Successful
responses to such a grand challenge, like the Digital
Patient, require this cross-disciplinary integration (An
& Cockrell, 2022; Grieves, 2019; P. Hunter et al.,
2002; Niarakis et al., 2022). Systems physiology
focuses on the function of interacting parts of the
system at the cell, tissue, organ and organ-system
scales, and is tightly coupled with structural
anatomical information (Sherwood, 2015). Systems
medicine is a subset of systems physiology that
addresses applications to clinical problems. Examples
include the application of the systems physiology
framework to develop quantitative understanding of
disease processes, leading to drug discovery, and to
the design of diagnostic tools (Tyson et al., 2001). A
subset of systems medicine that relies on individual
patient data or the data from a specific group of
similar patients is the emerging domain of
personalized medicine.
Realizing this goal requires the ability to make
accurate predictions about how a patient will react to
a treatment or no treatment; however, the previous
reductionist approach to science and modeling cannot
satisfy that need. Instead, the systems approach to
biological modeling is growing in importance as a
translational tool in clinical practice (Doyle III et al.,
2014). The explosion of data over the past twenty
years is providing novel opportunities to develop new
clinical treatments. New technologies such as DNA
sequencing, imaging, and proteomics provide
massive amounts of new information about the
human body. Further, these data can now be analyzed
at bulk or at the single-cell level, which has been
particularly useful to assess the tissue-level
heterogeneity in many diseases including cancer. The
ability to extract useful information from these data
has begun to lead to custom treatments for diseases,
such as cancer (Goldenberg et al., 2019; Khoo et al.,
2021), infectious diseases (Castiglioni et al., 2021; de
Fátima Cobre et al., 2021), diabetes (Kavakiotis et al.,
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
146
2017) and hematological and metabolic disorders (De
Bruyne et al., 2021). This will, in turn, help improve
the health of individuals by converting research
findings into diagnostic tools and procedures.
2 CHALLENGES IN
DEVELOPING A DIGITAL
PATIENT FRAMEWORK
A large-scale implementation of human health into a
digital format requires the construction and execution
of highly complex computer models composed of
several component submodels which span multiple
spatial and temporal scales (P. Hunter, 2020; Pan et
al., 2021). In addition, multi-scale data is needed to
build this computational representation of biological
processes of the whole-body under both disease-free
and diseased states. Thus, several pieces must be in
place to realize this interplay in a single
computational model. These pieces include
reductionist modeling at a variety of spatial and
temporal scales, the development of an ontology
allowing models to communicate with one another,
and finally, the creation of a top-level model that
allows reductionist models to be plugged in, creating
an integrated model framework for the testing and
generation of hypotheses. Different groups have
developed distinct philosophies for approaching these
challenges, but none has solved the problem
completely (Hussan et al., 2022). Several challenges
to building a Digital Patient framework have been
identified and are discussed below.
2.1 Lack of a High Throughput
Approach to Modeling
A Digital Patient describing the disease state and
treatment requires the development, validation, and
integration of numerous component submodels in the
context of a rapidly developing scientific
understanding of biological behaviors and continual
generation of new experimental and clinical data
(Laubenbacher et al., 2021; Masison et al., 2021).
Although individual laboratories around the world
may construct submodels, the development of a
comprehensive Digital Patient framework will
require modularity to ensure validation and
interoperability with one another. In this way, the
submodels will handle complexity with modules that
are properly annotated, curated, and documented, and
then linked semantically to establish more complex
models. Enabling such parallel development requires
a flexible simulation architecture that uses a multi-
scale map of all the relevant components of a patient’s
response to the disease, as well as responses to
available treatments s (An & Cockrell, 2022; Grande
Gutiérrez et al., 2021; Masison et al., 2021).
2.2 Lack of Model Reproducibility and
Transparency
It is essential that component models utilized in the
Digital Patient are reproducible and reusable.
Published models relevant to this discussion
demonstrate a lack of transparency in model
implementation (Baker, 2016; Fitzpatrick, 2019). Not
all published models are reproducible and hence not
reusable. Furthermore, a significant number of
published mathematical models are not experimentally
validated, making model extensions more difficult
(Blinov et al., 2021; Niarakis., et al., 2022).
2.3 Generation of Heterogeneous
High-Dimensional Data
Development of a multi-scale model of an organ
requires the collection of synchronous measurements
at multiple physiological scales. This includes omics
data from tissues and single cells, from diverse
experimental systems, including two-dimensional
(2D) and 3D cell cultures, in vivo and ex vivo animal
models, patients, and biophysical and structural data
from tissues and organs, combined with data
characterizing transport throughout the body.
Technologically this is very challenging if not
impossible (Vieira & Laubenbacher, 2022).
2.4 Lack of Standardization in Data
Collection and Model Specification
Ever growing uncoordinated and heterogeneous
formats of data that capture the various determinants
of our health from genomic sequences to behavioral
influence lack (Vieira & Laubenbacher, 2022).
Thus, the use of incompatible data structures
along with almost no standard model specification
and different software environments, make
distributed collaboration difficult (Lubbock & Lopez,
2021; Malik-Sheriff et al., 2020).
2.5 Lack of Effective Communication
and Collaboration Among
Biomedical Researchers
Building a useful Digital Patient requires improved
communication between clinicians, experimentalists,
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions
147
and modelers in order to create sufficiently credible
interchangeable computational models and/or tools
that have value in the clinic, such as mobile apps,
dedicated web pages and medical devices. Inevitably,
this has led to the inability to efficiently translate
basic science knowledge obtained from pre-clinical
studies into effective therapies (An & Cockrell, 2022;
Grieves, 2019. To date, relatively few clinical and
biological insights are currently translated into
computational models that could serve as building
blocks for the Digital Patient framework.
2.6 Health Information Management
Having clinical information in electronic form that is
computable has been a grand challenge for
biomedical informatics (Acosta et al., 2022).
Unfortunately, most health information still sits in
silos today and health information exchange for the
purpose of supporting care between organizations and
levels of care (e.g., hospital to primary care), has,
until very recently, been the exception rather than the
norm. It is fair to say that, to a large extent,
management of health information has encountered
the most variability when we consider other related
domains like bioinformatics, pharmaceutical research
and development and medical device technology in
the quest for integrated biomedicine. Post-hoc data
collection has been shown to be very expensive and
error-prone because data sources can be very diverse
and range from operational electronic health record
systems to well-structured longitudinal disease
registries and bio-banks. Therefore, capturing
structured and computable clinical data as part of
routine clinical practice is ideal as it may otherwise
impossible to capture the clinical context in which the
data were collected initially. In addition, effectively
managing the enormous amount of personalized data
requires the development of broadly accepted policies
addressing security, quality control, data mining, and
privacy protection. This represents another
fundamental challenge to completing the Digital
Patient.
2.7 Patient Data Management
Further complicating the construction of the Digital
Patient framework is the current lack of agreement on
how we categorize patient information. An individual
patient’s ideal data set includes molecular data,
clinical data, and social context data, and these data
sets are not often integrated in a manner that is
understandable or easily usable by patients or
healthcare providers or even by research domain
experts (Kondylakis et al., 2015). Developing a
consistent terminology and aggregation methodology
for this disparate data is therefore an additional
fundamental challenge to building the Digital Patient
framework. Privacy, synchronicity (the timeliness
with which models produce actionable information),
and clarity of data organization and analysis are also
fundamental challenges that must be addressed in
completing the Digital Patient platform. Recent
3 DEVELOPMENT IN BUILDING
THE DIGITAL PATIENT
FRAMEWORK
There are many collaborative and individual efforts
underway that address some of the issues important
to building out the Digital Patient framework.
Following is a summary of several past and current
efforts, and tools developed. This list is a selection
and not exhaustive.
Projects that contributed most significant progress
are the Physiome and the 12 Labours projects; briefly
described below, the Physiome Project is an umbrella
term that refers to human modelling with methods
accommodating cross-disciplinary science
(chemistry, biology, physics) and a breadth of
dimensional and temporal scale (sub-cellular to
organs, sub-microsecond to tens-of-years) (P. Hunter
et al., 2002). The International Union of Physiological
Sciences (IUPS) Physiome Project, established in
1993, focused on providing a “computational
framework for understanding human and other
eukaryotic physiology”. This effort resulted in
databases, markup languages, software for
computational models of cell functions, as well as
software for interacting with organ models, as was
described in P. J. Hunter & Borg, 2003; P. Hunter,
2004. The primary limitation with the Physiome
Project has been the lack of integration of the multiple
narrow-focused models that could, if successfully
integrated, lead to a comprehensive and integrative
model of human physiology (D. P. Nickerson et al.,
2015; Viceconti et al., 2008). Today this project has
been extended into the 12 Labours research effort
currently underway within the Auckland
Bioengineering Institute at the University of
Auckland in New Zealand.
3.1 12 Labours Project
Initiated by the Auckland Bioengineering Institute
(ABI), University of Auckland in New Zealand, the
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
148
intent of 12 Labours project is to extend the Physiome
Project to clinic and home-based healthcare
applications (P. Hunter, 2020). The focus and goal of
12 Labours is to create the infrastructure to integrate
multi-scale models into a whole-body computational
physiology system. This will include a platform for
precision medicine and sensor-based health
monitoring. Migrating the efforts from a research
focus to a clinical focus will necessarily require
changes in design and execution, e.g., model
reduction strategies must be utilized in order to
increase the speed at which models can be analyzed
and output a useful result. Additional foci include
coupling the physiome to body sensors for real-time
data exchange, an energy based mathematical
framework for understanding physiology, and a new
semantic approach to physics based multi-scale
modeling. In addition, with the goal of integrating
medical data with predictive physics modeling, this
project includes the development of workflow
management systems, the identification of
technologies for clinical translation of those
workflows, and the infrastructure for deploying those
workflows in a clinical environment. Some of the
suitable tools for workflow and data management
initially include Snake Make, NextFlow, iRods,
Pennsieve (Rajagopal et al., 2022).
Through the NIH Stimulation Peripheral Activity
to Relieve Conditions (SPARC) program, ABI has
been developing scaffolds of high-level descriptions
of organ anatomy on a 3D coordinate system
framework to relate multi-species organ models
(Osanlouy et al., 2021). These frameworks provide a
single common reference frame, upon which one can
register (or align) data across species, i.e., human,
mouse, pig. In other words, the framework is
consistent across multiple species, and the scaffolding
method facilitates cross-species comparisons as well
as the analysis of variation within a population.
Likewise, sub-scaffolds are used to define individual
characteristics within these scaffolds. The concept of
the whole-body scaffold reflects the use of this logic
for personalized models for virtual clinical trials via a
workflow in which organs and systems could be
assembled into whole body reference coordinates.
3.2 Computational Frameworks
Modularity is a particularly important consideration
when model dependencies are involved as the
introduction and absence of models can throw off the
entirety of a simulation. Similarly, multiple scales
require a framework with internal compensation to
account for varying definitions of, e.g., time.
Following are two examples of recent frameworks
that have been developed specifically to address these
concerns and that may serve to better light the way
forward for a Digital Patient framework. The first is
focused on medical digital twins and is a multi-
institutional effort of the Universities of Connecticut,
Michigan, and Florida. The second supports global
resource management and is out of Johns Hopkins
APL. Despite unrelated disciplines, the designs are
strikingly similar. If this review proves useful, there
may be others that can be reviewed in a similar
fashion. Following are two example computational
platforms for the integration of multiple, disparate
models.
A Modular Computational Framework for
Medical Digital Twins With a focus on digital
twins in a medical context, researchers from
University of Florida Health sought to
optimize modularity by eliminating the
potential pitfalls of model dependencies with
an open source, “digital twin” architecture
(Masison et al., 2021). Characterized as “hub
and spoke” and hereafter referred to as MCF
for Modular Computational Framework, it
includes four components: a runtime
configuration file, a global model state,
modules, and a simulation framework that
controls simulation runtime and provides data
structures and algorithms useful for the
development of modules. Modules are
individual models, which can be added and
taken away at will; each module must provide
a subclass that defines the data relevant to that
module to be stored in the model state, i.e., a
pure data API. As such, the model state houses
all potential model inputs and outputs,
providing an indirect connection between
modules. One model’s output is stored in the
model state and can only from there be
accessed as input by another model. This also
ensures that all model processes stay
contained within a particular module, thereby
providing a clear separation between the
model and the data. In one paper (Masison et
al., 2021), MCF was applied to an existing
dynamic computational model of the immune
response to a respiratory fungal pathogen to
illustrate the potential of extending it to a full
digital twin use case. The MCF simulator is
open-source and available at (Masison, Joseph
and Beezley, Jonathan and Mei, Yu and
Ribeiro, Henrique Assis Lopes and Knapp,
Adam C and Sordo Vieira, L and Adhikari,
Bandita and Scindia, Yogesh and Grauer,
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions
149
Michael and Helba, Brian and others, 2021).
Additional platform access information is
available in the reference.
System Integration with Multiscale
Networks (SIMoN) SIMoN was built to
realize the complex inter-relationships that
exist between the different facets of global
resource management (Hughes et al., 2020).
Each of these facets is a domain unto itself,
and models use their own geospatial,
temporal, and
other scales. SIMoN is an open-
source framework built to allow these
disparate models to be coupled. In the
referenced paper, they include climate,
population, and food-energy-water systems.
Specifically, SIMoN is used to “integrate
models and data from disparate domains by
predicting water availability in 2050, as it
depends on population growth, climate
change, and corresponding increases in
demand for thermoelectric cooling". While
SIMoN is not a biomedical use-case, the
challenges of integrating multiple distinct
models with different processes and scales
remain the same, requiring a framework that is
modular and extendable. Interestingly, SIMoN
employs a similar approach to the previous
example, with a “broker” construct taking the
place of the model state. Each model exists
inside of wrapper to standardize its interface
with the broker. The broker performs the
transformations necessary to reconcile the
varying geo-spatial scales of each model so
that those models can be integrated
seamlessly. In addition, it handles all data
inputs and outputs between models; like MCF,
this broker prevents direct dependencies of
one model on another. The only requirement
is that the models agree on the “scope” of a
given scale, e.g., the entirety of the contiguous
United States is the geo-spatial scope. The
scope can be subdivided by an individual
model as needed with the assumption that the
sum of all subdivisions exactly equals the
scope, with no overlap. It is not hard to
imagine how the same principle might be
applied to temporal and possibly other scales.
SIMoN is available at (Hughes, Marisa and
Kelbaugh, Michael and Campbell, Victoria
and Reilly, Elizabeth and Agarwala, Susama
and Wilt, Miller and Badger, Andrew and
Fuller, Evan and Ponzo, Dillon and Arevalo,
Ximena Calderon and others, 2020).
3.3 Collaborative Organizational
Efforts
The challenges of maximizing value from big data are
being addressed by the U.S. National Institutes of
Health’s (NIH) BD2K program, through the
European Union’s Horizon 2020 initiative, through
the European Big Data Value Association and
through various Chinese Ministry of Science and
Technology initiatives. In addition, the challenge of
encouraging consistency in terminology, ontology,
and registries is being addressed through the
International Health Terminology Standards
Development Organization, the Simulation Industry
Standards Organization and the NIH Data Discovery
Index Consortium. Moreover, model construction
and interoperability are the foci of the Physiome and
12 Labours Project, and of the researchers that are
involved in several organizations, including the
Virtual Physiological Human Institute (VPHi), the
U.S. Interagency Modeling and Analysis Group, and
its companion group, the Multi-Scale Modeling
Consortium. More such organizational efforts are
listed below.
Insigneo, based on the Institute for In Silico
Medicine, is a collaboration between the
University of Sheffield, UK and the Sheffield
Teaching Hospitals with a focus on clinical
translation of in silico medicine. The project
implements the ambition behind the European
VPHi program. This is the largest organization
in Europe dedicated to the development,
validation, and use of in silico medical
technology.
InSilco, dedicated to coronary artery disease,
is an international multidisciplinary
consortium focused on in silico trials for drug
eluting bioresorbable vascular scaffold design,
development, and evaluation.
Physiome Journal is dedicated to the
reproducibility and reusability of models.
Each article is linked to a primary publication
in a peer-reviewed journal and includes access
to the presented model itself. This effort is
supported by the IUPS, VPHi, the University
of Auckland, Digital Science, and more.
IMAG stands for Interagency Modeling and
Analysis Group (IMAG) based on NIH, USA,
holds multi-scale Modeling (MSM)
Consortium. Their goal includes supporting
research funding for modeling and analysis of
biomedical, biological, and behavioral
systems.
InSilicoWorld is a worldwide community of
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
150
practice working towards wider adoption of in
silico trials in the biomedical industry that is
currently led by Marco Viceconti (Viceconti et
al., 2008).
SimBios, lead by NIH and Stanford
University, is a center for physics-based
simulation of biological structures. It provides
SimTK, developed OpenSim, and publishes
Biomedical Computation Review. It has also
established an inventory of bio-sim tools
called SimBiome in 2017.
Tools that were developed by the organizational
efforts are listed below.
Markup Language Standards have been
developed to adequately describe physical and
physiological properties and processes
including CellML (Lloyd et al., 2004, FieldML
(Chang et al., 2007), TissueML (J.Q. et al.,
2004), AnatML (J.Q. et al., 2004), PhysioML
(J.Q. et al., 2004), SBML (Hucka et al., 2003).
DICOM: Digital Imaging and
Communications in Medicine (DICOM) is the
international standard for medical images and
related information (Lim & Zein, 2006). It
defines formats for medical images that can be
exchanged with the data and quality necessary
for clinical use.
OpenSim is an open-source simulation
software that was developed by the Stanford
National Center for Simulation in
Rehabilitation Research (Delp et al., 2007).
BioModels is a repository of mathematical
models representing biological systems and is
written in SBML (Li et al., 2010). Models
include signaling, protein-drug interactions,
metabolic pathways, epidemic, and more.
BioModels was developed by the Molecular
Networks team EMBL-EBI based in UK and
the SBML Team from Caltech, USA.
SimTK is a free biomedical project hosting
platform for the biomedical computation
community (Project-hosting platform for the
biomedical computation community, n.d.). It
provides an easy data sharing, shared resource
tracking and an infrastructure for community
connection and growth.
OpenEHR is an open standard specification
in health informatics that allows semantic
mapping annotations into EHR data storage
formats (openEHR, 2017). From 2010-2020,
OpenEHR has been deployed in Australia,
Brazil, Switzerland, Germany, Finland, UK,
Italy, Malta, Netherlands, Norway,
Philippines, Russia, Sweden, and Slovenia.
BioUML is web-based integrated
environment for systems biology and
collaborative analysis of biomedical data
(Russian Science Foundation, 2002). It was
funded and initiated by the Russian Science
Foundation in 2002. The initial goal of
BioUML was common purpose visual
language for formal descriptions of the
structure and function of biological systems.
The long-range plan is to be a computational
platform for the VPH and digital patient.
BioUML spans a comprehensive range of
capabilities, including access to biological
databases, powerful tools for systems biology
(visual modelling, simulation, parameters
fitting and analyses), a genome browser,
scripting (R, JavaScript), and a workflow
engine. The architecture is plugin-based.
Users create a visual representation of a model
and BioUML automatically generates the code
to simulate the model behavior. The current
version generates code in Java and uses its
own simulation engines. To support
collaborative work, there is a central
authentication and authorization system.
BioUML is open-source, in continued
development, and is actively used.
MLBox was developed by the collaboration
with the University of Miami’s Miller School
of Medicine, the Media and Information Lab
(MIL), Amazon Web Services, and the
OpeHealth Network to create MLBox.
MLBox is an automated machine learning
Python library to support the development of
digital twins that can take the place of patients
to better test treatments options (Das &
Cakmak, 2018). Inputs include data from
wearable sensors and other smart devices,
including biological, clinical, behavioral, and
environmental. These are collected over a
period of seven days and combined into a
“biological health algorithm”, which in turn
acts as a digital twin in treatment tests. The
MLBox platform is in Python and is device
agnostic, i.e., modular, which will allow input
types to adjusted based on individual needs
and constraints, as well as expanded in the
future as technology evolves. The initial focus
area is sleep apnea and its link to dementia and
heart disease and inputs include sleep patterns,
weight, environmental pressures, and stress
levels.
promor was developed by researchers at the
Eastern Virginia Medical School. Promor is an
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions
151
open-source R package that streamlines
biomarker discovery from proteomics data
and builds predictive models of disease
diagnosis and/or prognosis with top protein
biomarker candidates (Ranathunge et al.,
2023).
3.4 Other Collaborative Efforts
Biomedical research groups worldwide are
employing “digital twin” technologies to realize the
promise of personalized medicine. For example,
digital twins of the human heart can improve
diagnosis, prognosis, and therapies (Martinez-
Velazquez et al., 2019). Developers expect that
automated workflows for generating cardiac digital
twins could serve as a blueprint for the generation of
other types of medical digital twins (Corral-Acero et
al., 2020). Although medical digital twins are much
more difficult to develop than those for engineered
devices, they have begun to find applications in
improving human health. Examples include the
“artificial pancreas” for type 1 diabetes patients
(Breton et al., 2020; Brown et al., 2019; Kovatchev,
2019). In the artificial pancreas model, a template
mathematical model of human glucose metabolism
and a closed-loop control algorithm modeling insulin
delivery and data from an implanted glucose sensor
are customized into a patient-specific digital twin that
continuously calculates insulin needs and drives an
implanted pump that adjusts blood insulin
concentrations. Additionally, pediatric cardiac digital
twins combine template models of the heart with
patient-derived clinical measurements to optimize
some heart surgeries (Shang et al., 2019) and assess
the risk of thrombosis (Grande Gutiérrez et al., 2021;
Kondratova et al., 2019). The ARCHIMEDES
diabetes model expands these technologies by
including models not only of the progression of
diabetes within individual patients but also of medical
diagnosis, treatments, and the functioning of the health
care system that is providing the treatment (Du et al.,
2013; Eddy & Schlessinger, 2003a, 2003b).
More recently, the NIH Maximizing Investigators
Research Award (MIRA) was awarded to Dr. Tomas
Helikar, Professor of Biochemistry, University of
Nebraska, Lincoln, for the further development of a
virtual immune system. The virtual immune system is
meant to increase the understanding of immune
related diseases as well as to speed up drug
development and the time-to-market timeline. The
first MIRA award resulted in the successful modeling
of CD4+ T cells, which stimulate other cells to fight
pathogens. This model encompasses four
mathematical approaches, three spatial scales, and
multiple tissues involved in immune response. The
project established a method for computationally
connecting multiple scales of the immune system
(Wertheim et al., 2021. The goal of this second MIRA
award is to expand the model to include more types
of cells, molecules, genes, and organs.
A large part of the focus will be on computational
cost-effectiveness, improving the speed and
efficiency of the model’s algorithms.
In Europe, Neurotwin was initiated and funded by
the EU Horizon 2020 on January 1, 2021. It is
currently led by Neuroelectrics in Spain. It seeks to
predict the effects of non-invasive stimulation for
treatment of neurological disorders, e.g., Alzheimer’s
disease and epilepsy. Two proof-of-concept clinical
trials are planned for 2022 and 2023 in order to refine
the application of this stimulation technology for the
conditions of Alzheimer’s disease and epilepsy. If
successful, the condition use cases will be extended
to multiple sclerosis stroke rehabilitation, depression,
and the effects of psychedelics in the future.
Neurotwin combines 30 minutes of MRI and 10
minutes of EEG to create a personalized digital twin
that captures a brain’s electrical activity and
simulations the brain’s main parts, including the
scalp, skull, cerebrospinal fluid, and gray and white
matter. The digital twin also includes neural mass
models, or computational models of the average
behavior neurons using a map of neural connections
(connectome). This digital twin will be used to
optimize the stimulation position or locations and
strength of current via a headcap.
These examples illustrate how current digital
twins can operate in real time to maintain health
continuously, or they can be used off-line to design
personalized medical interventions.
3.5 Community Efforts
Community efforts such as the Systems Medicine
Disease Map Project, COVID-19 Disease Map
Project (Mazein et al., 2018; Ostaszewski et al., 2019;
Ostaszewski et al., 2020) and Computational
Modeling in Biology Network (COMBINE) are
working to build such infrastructure, although much
work needs to be done to adapt those for use in Digital
Patient framework. To this end, these groups have
built a large-scale data repository (Kondylakis et al.,
2017; Kondylakis et al., 2018; Kondylakis et al., 2015;
Kouroubali et al., 2019).
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
152
4 CONCLUSIONS AND FUTURE
DIRECTIONS
Data is everywhere now, being aggregated, analyzed,
and repackaged. We are in an era of Big Data, living
with the recognition that almost everything we do is
being captured as one or another type of data, with the
hope that all that data can be used to help us become
smarter, healthier, safer, and richer. We also
recognize that our privacy may be invaded and that
our risk for harm is increasing. It is in this broader
context that this article addresses one of the more
hopeful Big Data undertakings - that is, the
construction and deployment of the Digital Patient.
The capacity to measure one’s personal physiological
and social metrics, compare those metrics with the
metrics of millions of other humans, personalize
therapeutic interventions and measure the resulting
changes will ultimately realize the vision of
personalized medicine - wherein patients and their
providers will be able to detect disease at an earlier
age and provide optimal therapy based on the
characteristics of each individual and reduce adverse
responses to therapy. Similarly, pharmaceutical
companies will improve the process of drug discovery
and clinical trials. In this way, the healthcare
industry’s emphasis truly shifts from reaction to
disease to prevention of disease and promotion of
wellness. Implicit in this vision is the integration of a
sustained focus on improving the outcome measures
of healthcare-safety, effectiveness, patient-
centeredness timeliness, efficiency, and equity in
clinical practice. Underlying this focus is, of course,
the development and integration of multi-scale
models based on the understandings emerging from
systems biology.
While the application of physics-based models to
the Digital Patient are exciting and varied, several
substantial challenges face the community. The two
most critical needs are connecting the top-down and
bottom-up model approaches. Modeling languages
have been established separately, and so the
community must spend valuable time and effort
replicating work already done by other groups. This
represents a gross inefficiency in the development
process, and hampers cooperation between groups. It
is our belief that organizations such as, the industry-
academia-regulatory agency consortia Avicenna
(European Commission, University of Sheffield and
the consortium, 2013) and the Medical Device
Innovation Consortium (Research collaborations in
regulatory science, 2011) will provide the force to
consolidate these efforts into a unified whole. The
more troubling challenge is that of rigorous model
validation. The assumptions that underlie the model
induce a standard for evaluating the model. In the
case of a larger target such as tissue, organ, or an
individual, validation becomes a more difficult
concept to define. Intense inter-subject variations
exist in humans. A person even demonstrates
different physiological characteristics at different
ages, so the existence of a data set that represents a
target for validation is often in question. Population
modeling may be the key; by generating many
individuals, a class of subjects similar to a given
patient might be selected over a collection of
observable variables. Consideration of differences in
that population may suggest other observations to
make in the patient, establishing an iterative process
for matching an individual to a reasonable model.
This challenge is not unique to biological models; it
exists across all nonlinear dynamic models, and no
systematic solution has been accepted (Barlas, 1996;
Coveney & Fowler, 2005). That said, the potential for
improving the modeling of individual patients and the
strata of patients that is represented by the Digital
Patient is clearly worth pursuing.
Author Contributions: All authors have read and
agreed to the published version of the manuscript.
Funding: This work was supported by the Hampton
Roads Biomedical Research Consortium.
Institutional Review Board Statement: Not
applicable. Informed Consent Statement: Not
applicable.
Data Availability Statement: Not applicable.
Conflicts of Interest: The authors declare no conflict
of interest. The funders had no role in the design of
the study; in the collection, analyses, or interpretation
of data; in the writing of the manuscript; or in the
decision to publish the results.
REFERENCES
Acosta, J. N., Falcone, G. J., Rajpurkar, P., & Topol, E. J.
(2022). Multimodal biomedical ai. Nature Medicine, 28
(9), 1773–1784.
Alber, M., Buganza Tepole, A., Cannon, W. R., De, S.,
Dura-Bernal, S., Garikipati, K., Karniadakis, G.,
Lytton, W. W., Perdikaris, P., Petzold, L., et al. (2019).
Integrating machine learning and multiscale
modeling—perspectives, challenges, and opportunities
in the biological, biomedical, and behavioral sciences.
NPJ Digital Medicine, 2 (1), 1–11.
Altan-Bonnet, G., Mora, T., & Walczak, A. M. (2020).
Quantitative immunology for physicists. Physics
Reports, 849, 1–83.
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions
153
An, G., & Cockrell, C. (2022). Drug development digital
twins for drug discovery, testing and repurposing: A
schema for requirements and development. Frontiers in
Systems Biology, 2.
Auslander, D., Perelson, A., Clifford, G., & Oster, G.
(1972). On systems with coupled chemical reaction and
diffusion. The American Society of Mechanical
Engineers.
Baccam, P., Beauchemin, C., Macken, C. A., Hayden, F.
G., & Perelson, A. S. (2006). Kinetics of influenza a
virus infection in humans. Journal of virology, 80 (15),
7590–7599.
Baker, M. (2016). Reproducibility crisis. Nature, 533 (26),
353–66.
Barlas, Y. (1996). Formal aspects of model validity and
validation in system dynamics. System Dynamics
Review: The Journal of the System Dynamics Society,
12 (3), 183–210.
Bauer, A. L., Beauchemin, C. A., & Perelson, A. S. (2009).
Agent-based modeling of host–pathogen systems: The
successes and challenges. Information Sciences, 179
(10), 1379–1389.
Beauchemin, C. A., & Handel, A. (2011). A review of
mathematical models of influenza a infections within a
host or cell culture: Lessons learned and challenges
ahead. BMC Public Health,11 (1), 1–15.
Bertolaso, M. (2022). Understanding complexity in life
sciences. In Environmental alteration leads to human
disease (pp. 1–13). Springer.
Blinov, M. L., Gennari, J. H., Karr, J. R., Moraru, I. I.,
Nickerson, D. P., & Sauro, H. M. (2021). Practical
resources for enhancing the reproducibility of
mechanistic modeling in systems biology. Current
Opinion in Systems Biology, 27, 100350.
Boogerd, F. C., Bruggeman, F. J., Hofmeyr, J.-H. S., &
Westerhoff, H. V. (2007). Towards philosophical
foundations of systems biology: Introduction. In
Systems biology (pp. 3–19). Elsevier.
Breton, M. D., Kanapka, L. G., Beck, R. W., Ekhlaspour,
L., Forlenza, G. P., Cengiz, E., Schoelwer, M., Ruedy,
K. J., Jost, E., Carria, L., et al. (2020). A randomized
trial of closed-loop control in children with type 1
diabetes. New England Journal of Medicine, 383 (9),
836–845.
Bricmont, J. (2022). Conclusion: Statistical mechanics and
reductionism. In Making sense of statistical mechanics
(pp. 311–314). Springer.
Brigandt, I. (2013). Systems biology and the integration of
mechanistic explanation and mathematical explanation.
Studies in History and Philosophy of Science Part C:
Studies in History and Philosophy of Biological and
Biomedical Sciences, 44 (4), 477–492.
Brown, S. A., Kovatchev, B. P., Raghinaru, D., Lum, J. W.,
Buckingham, B. A., Kudva, Y. C., Laffel, L. M., Levy,
C. J., Pinsker, J. E., Wadwa, R. P., et al. (2019). Six-
month randomized, multicenter trial of closed-loop
control in type 1 diabetes. New England Journal of
Medicine, 381 (18), 1707–1717.
Castiglioni, I., Ippolito, D., Interlenghi, M., Monti, C. B.,
Salvatore, C., Schiaffino, S., Polidori, A., Gandola, D.,
Messa, C., & Sardanelli, F. (2021). Machine learning
applied on chest x-ray can aid in the diagnosis of covid-
19: A first experience from lombardy, italy. European
Radiology Experimental, 5 (1), 1–10.
Chang, D., Lovell, N. H., & Dokos, S. (2007). Field markup
language: Biological field representation in xml. 2007
29th Annual International Conference of the IEEE
Engineering in Medicine and Biology Society, 402–
405.
Combs, C. D., Sokolowski, J. A., & Banks, C. M. (2015).
The digital patient: Advancing healthcare, research, and
education. John Wiley & Sons.
Combs, C. D. (2017). Developing a framework for multi-
scale modeling of the digital patient. International
Journal of Privacy and Health Information
Management (IJPHIM), 5 (2), 21–33.
Combs, C. D., & Combs, W. C. (2014). The digital patient:
An emerging platform for health analytics. Wiley
StatsRef: Statistics Reference Online, 1–9.
Corral-Acero, J., Margara, F., Marciniak, M., Rodero, C.,
Loncaric, F., Feng, Y., Gilbert, A., Fernandes, J. F.,
Bukhari, H. A., Wajdan, A., et al. (2020). The ‘digital
twin’ to enable the vision of precision cardiology.
European Heart Journal, 41 (48), 4556–4564.
Costello, Z., & Martin, H. G. (2018). A machine learning
approach to predict metabolic pathway dynamics from
time-series multiomics data. NPJ Systems Biology and
Applications, 4 (1), 1–14.
Coveney, P. V., & Fowler, P. W. (2005). Modelling
biological complexity: A physical scientist’s
perspective. Journal of the Royal Society Interface, 2
(4), 267–280.
Das, S., & Cakmak, U. M. (2018). Hands-on automated
machine learning: A beginner’s guide to building
automated machine learning systems using automl and
python. Packt Publishing Ltd.
De Bruyne, S., Speeckaert, M. M., Van Biesen, W., &
Delanghe, J. R. (2021). Recent evolutions of machine
learning applications in clinical laboratory medicine.
Critical Reviews in Clinical Laboratory Sciences, 58
(2), 131–152.
de Fátima Cobre, A., Stremel, D. P., Noleto, G. R., Fachi,
M. M., Surek, M., Wiens, A., Tonin, F. S., & Pontarolo,
R. (2021). Diagnosis and prediction of covid-19
severity: Can biochemical tests and machine learning
be used as prognostic indicators? Computers in Biology
and Medicine, 134, 104531.
Delp, S. L., Anderson, F. C., Arnold, A. S., Loan, P., Habib,
A., John, C. T., Guendelman, E., & Thelen, D. G.
(2007). Opensim: Open-source software to create and
analyze dynamic simulations of movement. IEEE
Transactions on Biomedical Engineering, 54 (11),
1940–1950.
Dıaz-Zuccarini, V., Alimohammadi, M., & Pichardo-
Almarza, C. (2015). Reflecting on discipulus and
remaining challenges. The Digital Patient: Advancing
Healthcare, Research, and Education, 15.
Doyle III, F. J., Huyett, L. M., Lee, J. B., Zisser, H. C., &
Dassau, E. (2014). Closed-loop artificial pancreas
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
154
systems: Engineering the algorithms. Diabetes Care, 37
(5), 1191–1197.
Du, P., O’Grady, G., Gao, J., Sathar, S., & Cheng, L. K.
(2013). Toward the virtual stomach: Progress in
multiscale modeling of gastric electrophysiology and
motility. Wiley Interdisciplinary Reviews: Systems
Biology and Medicine, 5 (4), 481–493.
Eddy, D. M., & Schlessinger, L. (2003a). Archimedes: A
trial-validated model of diabetes. Diabetes Care, 26
(11), 3093–3101.
Eddy, D. M., & Schlessinger, L. (2003b). Validation of the
archimedes diabetes model. Diabetes Care, 26 (11),
3102–3110.
European Commission, University of Sheffield and the
consortium. (2013). Avicenna: A strategy for in silico
clinical trials [http://avicenna-isct.org/].
Fitzgerald, J. B., Schoeberl, B., Nielsen, U. B., & Sorger, P.
K. (2006). Systems biology and combination therapy in
the quest for clinical efficacy. Nature Chemical
Biology, 2 (9), 458–466.
Fitzpatrick, B. G. (2019). Issues in reproducible simulation
research. Bulletin of Mathematical Biology, 81 (1), 1–
6.
Giansanti, D. (2022). Artificial intelligence in public health:
Current trends and future possibilities. Gold, K.,
Gaharwar, A. K., & Jain, A. (2019). Emerging trends in
multiscale modeling of vascular pathophysiology:
Organ-on-a-chip and 3d printing. Biomaterials, 196, 2–
17.
Goldenberg, S. L., Nir, G., & Salcudean, S. E. (2019). A
new era: Artificial intelligence and machine learning in
prostate cancer. Nature Reviews Urology, 16 (7), 391–
403.
Grande Gutrrez, N., Alber, M., Kahn, A. M., Burns, J. C.,
Mathew, M., McCrindle, B. W., & Marsden, A. L.
(2021). Computational modeling of blood component
transport related to coronary artery thrombosis in
kawasaki disease. PLoS Computational Biology, 17 (9),
e1009331.
Grieves, M. W. (2019). Virtually intelligent product
systems: Digital and physical twins.
Harline, K., Martınez-omez, J., Specht, C. D., & Roeder,
A. H. (2021). A life cycle for modeling biology at
different scales. Frontiers in Plant Science, 1724.
Hartman, P., Beckman, K., Silverstein, K., Yohe, S.,
Schomaker, M., Henzler, C., Onsongo, G., Lam, H. C.,
Munro, S., Daniel, J., et al. (2019). Next generation
sequencing for clinical diagnostics: Five year
experience of an academic laboratory. Molecular
Genetics and Metabolism Reports, 19, 100464.
Hatzikirou, H., Chauviere, A., Bauer, A. L., Leier, A.,
Lewis, M. T., Macklin, P., Marquez-Lago, T. T.,
Bearer, E. L., & Cristini, V. (2012). Integrative physical
oncology. Wiley Interdisciplinary Reviews: Systems
Biology and Medicine, 4 (1), 1–14.
Hester, R. L., Iliescu, R., Summers, R., & Coleman, T. G.
(2011). Systems biology and integrative physiological
modelling. The Journal of Physiology, 589 (5), 1053–
1060.
Hucka, M., Finney, A., Sauro, H. M., Bolouri, H., Doyle, J.
C., Kitano, H., Arkin, A. P., Bornstein, B. J., Bray, D.,
Cornish-Bowden, A., et al. (2003). The systems biology
markup language (sbml): A medium for representation
and exchange of biochemical network models.
Bioinformatics, 19 (4), 524–531.
Hughes, M., Kelbaugh, M., Campbell, V., Reilly, E.,
Agarwala, S., Wilt, M., Badger, A., Fuller, E., Ponzo,
D., Arevalo, X. C., et al. (2020). System integration
with multiscale networks (simon): A modular
framework for resource management models. 2020
Winter Simulation Conference (WSC), 656–667.
Hughes, Marisa and Kelbaugh, Michael and Campbell,
Victoria and Reilly, Elizabeth and Agarwala, Susama
and Wilt, Miller and Badger, Andrew and Fuller, Evan
and Ponzo, Dillon and Arevalo, Ximena Calderon and
others. (2020). System integration with multiscale
networks [https://github.com/JHUAPL/SIMoN].
Hunter, P. (2020). Modeling framework for computational
physiology. Encyclopedia of Continuum Mechanics,
1691–1702.
Hunter, P., Robbins, P., & Noble, D. (2002). The iups
human physiome project. Pfl¨ugers Archiv,445 (1), 1–
9.
Hunter, P. J., & Borg, T. K. (2003). Integration from
proteins to organs: The physiome project. Nature
Reviews Molecular Cell Biology, 4 (3), 237–243.
Hunter, P. (2004). The iups physiome project: A framework
for computational physiology. Progress in Biophysics
and Molecular Biology, 85 (2-3), 551–569.
Hussan, J. R., Trew, M. L., & Hunter, P. J. (2022).
Simplifying the process of going from cells to tissues
using statistical mechanics. Frontiers in physiology,
279.
Iglesias, P. A., & Ingalls, B. P. (2010). Control theory and
systems biology. MIT press.
Ingalls, B. P. (2013). Mathematical modeling in systems
biology: An introduction. MIT press.
J.Q., D., Q., L., & Q.M., L. (2004). Java and xml based
graphic software package for modeling biochemical
and gene regulatory networks. Computational
Engineering Application, 40 (6), 202–204.
Kamerlin, S. C. L., & Warshel, A. (2011). Multiscale
modeling of biological functions. Physical Chemistry
Chemical Physics, 13 (22), 10401–10411.
Karr, J., Malik-Sheriff, R. S., Osborne, J., Gonzalez-Parra,
G., Forgoston, E., Bowness, R., Liu, Y., Thompson, R.,
Garira, W., Barhak, J., et al. (2022). Model integration
in computational biology: The role of reproducibility,
credibility and utility. Frontiers in Systems Biology, 2.
Kavakiotis, I., Tsave, O., Salifoglou, A., Maglaveras, N.,
Vlahavas, I., & Chouvarda, I. (2017). Machine learning
and data mining methods in diabetes research.
Computational and Structural Biotechnology Journal,
15, 104–116.
Kennedy, M. C., & O’Hagan, A. (2001). Bayesian
calibration of computer models. Journal of the Royal
Statistical Society: Series B (Statistical Methodology),
63 (3), 425–464.
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions
155
Khoo, A., Liu, L. Y., Nyalwidhe, J. O., Semmes, O. J.,
Vesprini, D., Downes, M. R., Boutros, P. C., Liu, S. K.,
& Kislinger, T. (2021). Proteomic discovery of non-
invasive biomarkers of localized prostate cancer using
mass spectrometry. Nature Reviews Urology, 18 (12),
707–724.
Kitano, H. (2001). Foundations of systems biology. The
MIT Press Cambridge, Massachusetts London,
England.
Klipp, E., Herwig, R., Kowald, A., Wierling, C., &
Lehrach, H. (2005). Systems biology in practice:
Concepts, implementation and application. John Wiley
& Sons.
Klipp, E., Liebermeister, W., Wierling, C., & Kowald, A.
(2016). Systems biology: A textbook. John Wiley &
Sons.
Kohl, P., Crampin, E. J., Quinn, T., & Noble, D. (2010).
Systems biology: An approach. Clinical Pharmacology
& Therapeutics, 88 (1), 25–33.
Kondratova, M., Czerwinska, U., Sompairac, N.,
Amigorena, S. D., Soumelis, V., Barillot, E., Zinovyev,
A., & Kuperstein, I. (2019). A multiscale signalling
network map of innate immune response in cancer
reveals cell heterogeneity signatures. Nature
Communications, 10 (1), 1–13.
Kondylakis, H., Bucur, A., Dong, F., Renzi, C., Manfrinati,
A., Graf, N., Hoffman, S., Koumakis, L., Pravettoni, G.,
Marias, K., et al. (2017). Imanagecancer: Developing a
platform for empowering patients and strengthening
self-management in cancer diseases. 2017 IEEE 30th
International Symposium on Computer-Based Medical
Systems (CBMS), 755–760.
Kondylakis, H., Koumakis, L., Tsiknakis, M., & Marias, K.
(2018). Implementing a data management
infrastructure for big healthcare data. 2018 IEEE
EMBS International Conference on Biomedical &
Health Informatics (BHI), 361–364.
Kondylakis, H., Spanakis, E. G., Sfakianakis, S., Sakkalis,
V., Tsiknakis, M., Marias, K., Zhao, X., Yu, H. Q., &
Dong, F. (2015). Digital patient: Personalized and
translational data management through the
MyHealthAvatar EU project. 2015 37th Annual
International Conference of the IEEE Engineering in
Medicine and Biology Society (EMBC), 1397–1400.
Kouroubali, A., Koumakis, L., Kondylakis, H., &
Katehakis, D. G. (2019). An integrated approach
towards developing quality mobile health apps for
cancer. In Mobile health applications for quality
healthcare delivery (pp. 46–71). IGI Global.
Kovatchev, B. (2019). A century of diabetes technology:
Signals, models, and artificial pancreas control. Trends
in Endocrinology & Metabolism, 30 (7), 432–444.
Kuijper, H. (2022). Reductionism or holism? In
Comprehending the complexity of countries (pp. 247–
273). Springer.
Laubenbacher, R., Sluka, J. P., & Glazier, J. A. (2021).
Using digital twins in viral infection.Science, 371
(6534), 1105–1106.
Li, C., Donizelli, M., Rodriguez, N., Dharuri, H., Endler,
L., Chelliah, V., Li, L., He, E., Henry, A., Stefan, M. I.,
et al. (2010). Biomodels database: An enhanced,
curated and annotated resource for published
quantitative kinetic models. BMC Systems Biology, 4
(1), 1–14.
Likić, V. A., McConville, M. J., Lithgow, T., & Bacic, A.
(2010). Systems biology: The next frontier for
bioinformatics. Advances in Bioinformatics, 2010.
Lim, J., & Zein, R. (2006). The digital imaging and
communications in medicine (dicom): Description,
structure and applications. In Rapid prototyping (pp.
63–86). Springer.
Linka, K., Peirlinck, M., & Kuhl, E. (2020). The
reproduction number of covid-19 and its correlation
with public health interventions. Computational
Mechanics, 66 (4), 1035–1050.
Lloyd, C. M., Halstead, M. D., & Nielsen, P. F. (2004).
Cellml: Its future, present and past. Progress in
Biophysics and Molecular Biology, 85 (2-3), 433–450.
Lubbock, A. L., & Lopez, C. F. (2021). Programmatic
modeling for biological systems. Current Opinion in
Systems Biology, 27, 100343.
Maass, W., Parsons, J., Purao, S., Storey, V. C., & Woo, C.
(2018). Data-driven meets theory-driven research in the
era of big data: Opportunities and challenges for
information systems research. Journal of the
Association for Information Systems, 19 (12), 1.
Malik-Sheriff, R. S., Glont, M., Nguyen, T. V., Tiwari, K.,
Roberts, M. G., Xavier, A., Vu, M. T., Men, J., Maire,
M., Kananathan, S., et al. (2020). Biomodels—15 years
of sharing computational models in life science.
Nucleic Acids Research, 48 (D1), D407–D415.
Martinez-Velazquez, R., Gamez, R., & El Saddik, A.
(2019). Cardio twin: A digital twin of the human heart
running on the edge. 2019 IEEE International
Symposium on Medical Measurements and
Applications (MeMeA), 1–6.
Masison, J., Beezley, J., Mei, Y., Ribeiro, H. A. L., Knapp,
A. C., Sordo Vieira, L., Adhikari, B., Scindia, Y.,
Grauer, M., Helba, B., et al. (2021). A modular
computational framework for medical digital twins.
Proceedings of the National Academy of Sciences, 118
(20), e2024287118.
Masison, Joseph and Beezley, Jonathan and Mei, Yu and
Ribeiro, Henrique Assis Lopes and Knapp, Adam C and
Sordo Vieira, L and Adhikari, Bandita and Scindia,
Yogesh and Grauer, Michael and Helba, Brian and
others. (2021). A modular computational framework
for medical digital twins [https://github.com/
NutritionalLungImmunity/nlisim].
Mazein, A., Ostaszewski, M., Kuperstein, I., Watterson, S.,
Le Nov`ere, N., Lefaudeux, D., De Meulder, B., Pellet,
J., Balaur, I., Saqi, M., et al. (2018). Systems medicine
disease maps: Community-driven comprehensive
representation of disease mechanisms. NPJ Systems
Biology and Applications, 4 (1), 1–10.
Mazzocchi, F. (2008). Complexity in biology: Exceeding
the limits of reductionism and determinism using
complexity theory. EMBO Reports, 9 (1), 10–14.
Meier-Schellersheim, M., Fraser, I. D., & Klauschen, F.
(2009). Multiscale modeling for biologists. Wiley
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
156
Interdisciplinary Reviews: Systems Biology and
Medicine, 1 (1), 4–14.
Morchio, R. (1991). Reductionism in biology. In The
problem of reductionism in science (pp. 149–160).
Springer.
Muzio, G., O’Bray, L., & Borgwardt, K. (2021). Biological
network analysis with deep learning. Briefings in
Bioinformatics, 22 (2), 1515–1530.
Narkhede, S. M., Luther, L., Raugh, I. M., Knippenberg, A.
R., Esfahlani, F. Z., Sayama, H., Cohen, A. S.,
Kirkpatrick, B., & Strauss, G. P. (2022). Machine
learning identifies digital phenotyping measures most
relevant to negative symptoms in psychotic disorders:
Implications for clinical trials. Schizophrenia Bulletin,
48 (2), 425–436.
Niarakis, A., Waltemath, D., Glazier, J., Schreiber, F.,
Keating, S. M., Nickerson, D., Chaouiya, C., Siegel, A.,
No¨el, V., Hermjakob, H., et al. (2022). Addressing
barriers in comprehensiveness, accessibility,
reusability, interoperability and reproducibility of
computational models in systems biology. Briefings in
Bioinformatics.
Nickerson, D. P., Atalag, K., de Bono, B., & Hunter, P. J.
(2015). The physiome project, openehr, archetypes, and
the digital patient. The Digital Patient: Advancing
Healthcare, Research, and Education, John Wiley &
Sons, 101–124.
Nickerson, D. P., Lundeng˚ard, K., Watts, J., Porter, S., Yu,
T., Nielsen, P., & Hunter, P. (2020). Physiome:
Encouraging reproducible and fair computational
modelling. The FASEB Journal, 34 (S1), 1–1.
openEHR. (2017). An open domain-driven platform for
developing flexible e-health systems.
Osanlouy, M., Bandrowski, A., De Bono, B., Brooks, D.,
Cassar`a, A. M., Christie, R., Ebrahimi, N., Gillespie,
T., Grethe, J. S., Guercio, L. A., et al. (2021). The sparc
drc: Building a resource for the autonomic nervous
system community. Frontiers in Physiology, 12.
Ostaszewski, M., Gebel, S., Kuperstein, I., Mazein, A.,
Zinovyev, A., Dogrusoz, U., Hasenauer, J., Fleming, R.
M., Le Novere, N., Gawron, P., et al. (2019).
Community-driven roadmap for integrated disease
maps. Briefings in Bioinformatics, 20 (2), 659–670.
Ostaszewski, M., Mazein, A., Gillespie, M. E., Kuperstein,
I., Niarakis, A., Hermjakob, H., Pico, A. R.,
Willighagen, E. L., Evelo, C. T., Hasenauer, J., et al.
(2020). COVID-19 disease map, building a
computational repository of SARS-CoV-2 virus-host
interaction mechanisms. Scientific Data, 7 (1), 1–4.
Oster, G. F., & Perelson, A. S. (1973). Systems, circuits and
thermodynamics. Israel Journal of Chemistry, 11 (2-3),
445–478.
Pan, M., Gawthrop, P. J., Cursons, J., & Crampin, E. J.
(2021). Modular assembly of dynamic models in
systems biology. PLoS Computational Biology, 17
(10), e1009513.
Parodi, V. A. (2015). The digital patient: Changing the
paradigm of healthcare and impacting medical research
and education. The Digital Patient: Advancing
Healthcare, Research, and Education, 273–88.
Peng, G. C., Alber, M., Buganza Tepole, A., Cannon, W.
R., De, S., Dura-Bernal, S., Garikipati, K., Karniadakis,
G., Lytton, W. W., Perdikaris, P., et al. (2021).
Multiscale modeling meets machine learning: What can
we learn? Archives of Computational Methods in
Engineering, 28 (3), 1017–1037.
Perelson, A. S., Neumann, A. U., Markowitz, M., Leonard,
J. M., & Ho, D. D. (1996). Hiv-1 dynamics in vivo:
Virion clearance rate, infected cell life-span, and viral
generation time. Science, 271 (5255), 1582–1586.
Perelson, A. S., & Weisbuch, G. (1997). Immunology for
physicists. Reviews of Modern Physics, 69 (4), 1219.
Pinky, L., Burke, C. W., Russell, C. J., & Smith, A. M.
(2021). Quantifying dose-, strain-, and tissue-specific
kinetics of parainfluenza virus infection. PLoS
Computational Biology, 17 (8), e1009299.
Pinky, L., & Dobrovolny, H. M. (2017). The impact of cell
regeneration on the dynamics of viral coinfection.
Chaos: An Interdisciplinary Journal of Nonlinear
Science, 27 (6), 063109.
Project-hosting platform for the biomedical computation
community. (n.d.). Web-based integrated environment
for systems biology [https://simtk.org/
whatIsSimtk.php].
Pruett, W. A., & Hester, R. L. (2015). Physics-based
modeling for the physiome. The Digital Patient:
Advancing Healthcare, Research, and Education, 127.
Radhakrishnan, R. (2020). Multiscale modeling:
Foundations, historical milestones, current status, and
future prospects. Authorea Preprints.
Radhakrishnan, R. (2021). A survey of multiscale
modeling: Foundations, historical milestones, current
status, and future prospects. AIChE Journal, 67 (3),
e17026.
Rajagopal, V., Arumugam, S., Hunter, P., Khadangi, A.,
Chung, J., & Pan, M. (2022). The cell physiome: What
do we need in a computational physiology framework
for predicting single cell biology? arXiv preprint
arXiv:2202.13282.
Ranathunge, C., Patel, S. S., Pinky, L., Correll, V. L., Chen,
S., Semmes, O. J., Armstrong, R. K., Combs, C. D., &
Nyalwidhe, J. O. (2023). promor: a comprehensive R
package for label-free proteomics data analysis and
predictive modeling [vbad025]. Bioinformatics
Advances,3 (1). https://doi.org/10.1093/bioadv/
vbad025
Research collaborations in regulatory science. (2011).
Medical device innovation consortium [http://mdic.
org/].
Russian Science Foundation. (2002). Web-based integrated
environment for systems biology [https://bio-store.org].
Schaffner, K. F. (1976). Reductionism in biology:
Prospects and problems. PSA 1974, 613–632.
Sego, T., Mochan, E. D., Ermentrout, G. B., & Glazier, J.
A. (2022). A multiscale multicellular spatiotemporal
model of local influenza infection and immune
response. Journal of Theoretical Biology, 532, 110918.
Shang, J. K., Esmaily, M., Verma, A., Reinhartz, O.,
Figliola, R. S., Hsia, T.-Y., Feinstein, J. A., & Marsden,
A. L. (2019). Patient-specific multiscale modeling of
Developing a Framework for Multi-Scale Modeling of the Digital Patient: Insights from Current Status and Future Directions
157
the assisted bidirectional glenn. The Annals of Thoracic
Surgery, 107 (4), 1232–1239.
Sherwood, L. (2015). Human physiology: From cells to
systems. Cengage learning. Smith, A. M. (2018).
Validated models of immune response to virus
infection. Current Opinion in Systems Biology, 12, 46–
52.
Talbot, S., Foster, S. L., & Woolf, C. J. (2016).
Neuroimmunity: Physiology and pathology. Annual
Review of Immunology, 34, 421–447.
Teichert, G. H., Natarajan, A. R., Van der Ven, A., &
Garikipati, K. (2019). Machine learning materials
physics: Integrable deep neural networks enable scale
bridging by learning free energy functions. Computer
Methods in Applied Mechanics and Engineering, 353,
201–216.
Tolk, A., Balci, O., Combs, C. D., Fujimoto, R., Macal, C.
M., Nelson, B. L., & Zimmerman, P. (2015). Do we
need a national research agenda for modeling and
simulation? 2015 Winter Simulation Conference
(WSC), 2571–2585.
Tyson, J. J., Chen, K., & Novak, B. (2001). Network
dynamics and cell physiology. Nature Reviews
Molecular Cell Biology, 2 (12), 908–916.
Viceconti, M., Clapworthy, G., & Jan, S. V. S. (2008). The
virtual physiological human—a european initiative for
in silico human modelling—. The Journal of
Physiological Sciences, 0810200082–0810200082.
Vieira, L. S., & Laubenbacher, R. C. (2022). Computational
models in systems biology: Standards, dissemination,
and best practices. Current Opinion in Biotechnology,
75, 102702.
Wertheim, K. Y., Puniya, B. L., La Fleur, A., Shah, A. R.,
Barberis, M., & Helikar, T. (2021). A multi-approach
and multi-scale platform to model cd4+ t cells
responding to infections. PLoS Computational Biology,
17 (8), e1009209.
Woese, C. R. (2004). A new biology for a new century.
Microbiology and Molecular Biology Reviews, 68 (2),
173–186.
SIMULTECH 2023 - 13th International Conference on Simulation and Modeling Methodologies, Technologies and Applications
158