You are on page 1of 17

The Emerging Paradigm of Network Medicine in the Study of Human Disease

Stephen Y. Chan and Joseph Loscalzo

Circ Res. 2012;111:359-374


doi: 10.1161/CIRCRESAHA.111.258541
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2012 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/111/3/359

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the
Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information
about this process is available in the Permissions and Rights Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Circulation Research is online at:


http://circres.ahajournals.org//subscriptions/

Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014


Review

The Emerging Paradigm of Network Medicine in the Study


of Human Disease
Stephen Y. Chan, Joseph Loscalzo

Abstract: The molecular pathways that govern human disease consist of molecular circuits that coalesce into
complex, overlapping networks. These network pathways are presumably regulated in a coordinated fashion, but
such regulation has been difficult to decipher using only reductionistic principles. The emerging paradigm of
“network medicine” proposes to utilize insights garnered from network topology (eg, the static position of
molecules in relation to their neighbors) as well as network dynamics (eg, the unique flux of information through
the network) to understand better the pathogenic behavior of complex molecular interconnections that
traditional methods fail to recognize. As methodologies evolve, network medicine has the potential to capture the
molecular complexity of human disease while offering computational methods to discern how such complexity
controls disease manifestations, prognosis, and therapy. This review introduces the fundamental concepts of
network medicine and explores the feasibility and potential impact of network-based methods for predicting
individual manifestations of human disease and designing rational therapies. Wherever possible, we emphasize
the application of these principles to cardiovascular disease. (Circ Res. 2012;111:359-374.)
Key Words: network medicine ! systems biology ! cardiovascular disease ! systems pharmacology

F or centuries, reductionistic thinking has dominated West-


ern scientific theory and has been rigorously applied to
biomedicine. This construct argues that crucial biological
medicine, a “node” may represent a biological factor (eg,
gene, chromosome, metabolite, noncoding RNA, or even a
specific disease/phenotype). “Edges” may represent physical
factors operate in simple mechanistic association to control interactions, transcriptional induction, enzyme activation,
disease pathobiology. Consequently, current classification of enzymatic conversion of one metabolite to another, or even
disease phenotype (pathophenotype) is the result of inductive shared genes/traits among distinct disease manifestations. As
generalization from clinicopathological evidence founded on a result of rapidly progressing technology and high-
the law of parsimony. This paradigm has been helpful to throughput data sets, the first representations of a compre-
clinicians as it formalizes syndromic patterns that limit the hensive network, or “interactome,” of relevant functional
number of potential pathophenotypes. While quite useful in molecular interactions in human tissue is now possible.
an earlier era, classifying disease in this way overgeneralizes Currently, data accumulation for network construction has
pathophenotypes and does not usually consider individual- primarily relied on an integration of genetic studies (linkage
ized nuances in disease expression, susceptibility states, or analyses and genome-wide association studies, GWAS), so-
preclinical disease manifestations. Underlying those varia- called high-throughput –OMIC analyses (genomic sequenc-
tions in predisposition to disease and subsequent disease ing, transcriptional expression analyses, proteomic analyses,
manifestation is the competing concept that complex biolog- etc), and biochemical studies (ie, metabolomic analyses),
ical processes driving human disease rarely result from an coupled with cellular, physiological, and clinical data.
abnormality in a single molecular effector. Rather, they are As a result, application of network theory has begun to
nearly always the net result of multiple pathobiological permeate the scientific study of a large number of defined
pathways that interact through an interconnected network. biomedical systems, ranging from simple signaling circuits to
Network theory offers a tractable structure for distilling complex human diseases, such as cancer. The science of
relevant insight from the growing sets of molecular “-omics” studying disease network behavior is just now emerging and
data. In its simplest form, a network can be generated by has been termed “network medicine” (Figure 1B).1 In this
mapping a set of molecular entities, called “nodes,” with their review, we will define the challenges of building the current
positions based on their functional interconnections, called human “interactome” and the fundamental principles of
“links” or “edges,” with one another (Figure 1A). In network biological network theory in medicine. In doing so, we will

Original received April 5, 2012; revision received June 11, 2012; accepted June 19, 2012. In May 2012, the average time from submission to first
decision for all original research papers submitted to Circulation Research was 12.0 days.
From the Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA.
Correspondence to Joseph Loscalzo, MD, PhD, Department of Medicine, Brigham and Women’s Hospital, 75 Francis St, Boston, MA 02115. E-mail
jloscalzo@partners.org
© 2012 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.111.258541

359
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
360 Circulation Research July 20, 2012

for the analysis of transcriptional network dynamics, respec-


Non-standard Abbreviations and Acronyms
tively.6 “Sensitivity analysis,” or so-called “metabolic control
CALM1 calmodulin analysis” (MCA), is used to compute sensitivity coefficients,
CETP cholesterol ester transfer protein which generally reflect the ratios between the change in the
ChIP-ChIP chromatin immunoprecipitation followed by microarray biochemical network behavior (as monitored by some mea-
analysis sure of global system behavior) and the perturbation on
ChIP-Seq ChIP followed by sequencing specific system nodes.3,4 Resulting parameters with large
FDA-AERS Adverse Events Reporting System of the FDA sensitivity magnitude are deemed to be biologically important
FDA Food and Drug Administration and hence considered to be the controlling factors in the
GWAS genome-wide association studies functional regulation of a system.7 Various software packages
HCMV human cytomegalovirus for kinetic modeling of biochemical networks are available
LCMS likelihood-based causality model selection test and continue to grow in number.8 Although most software
LQTS long-QT syndrome shares a common core of functionality, the specific capabil-
MCA metabolic control analysis ities and user interfaces of different packages differ with
mRNA messenger RNA respect to their functionality, reliability, efficiency, user-
miRNA microRNA friendliness, and compatibility.
ODEs ordinary differential equations
Validation of the predictions arising from such MCA have
PPAR peroxisome proliferator-activated receptor
mostly centered on relatively narrow biochemical networks
linked by defined rate and time constants that are consistent
PPI protein-protein interactions
among a variety of perturbations. Although in many biolog-
QTL quantitative trait loci
ical circumstances such rate constants fluctuate and depend
on regulation of the catalyzing enzyme, the tight homeostatic
regulation that is seen in many biochemical networks, in part,
allows for consistent approximations of rate constants with-
explore how the study of network topology and network dynam- out substantially altering the network output. Moreover,
ics can accelerate discovery of novel disease genes and path- probabilistic estimations of dynamically changing rate con-
ways, offer new insights into the systems-wide effects of drug stants have been attempted.9 Such methodologies have vali-
therapy, and reshape disease classification. In the course of this dated important molecular features in metabolic and signaling
presentation, we will highlight key studies, cardiovascular and cascades at the single cell level. Consequently, one of the
otherwise, that have successfully integrated network theory with uses of MCA is to infer the importance of cellular processes
experimental validation and thus have demonstrated the power or pathways and to provide mechanistic explanations for
of these approaches for enhancing our understanding of molec- biological behavior.10 –14 As the technology improves for
ular and cellular interconnectedness and its potential regulation detecting dynamic changes in metabolites via high through-
in the control of disease phenotype. put metabolomics in a variety of clinically relevant scenar-
ios,15 the potential capabilities of such biochemical kinetic
Modeling of Biochemical Networks modeling should continue to expand.
Although most areas of modern medicine have relied nearly
exclusively on reductionist models of human pathogenesis, Modeling of Transcriptional Gene
efforts to establish and apply a detailed understanding of Regulatory Networks
networks for predicting “downstream” effects on cellular or The success of such biochemical modeling strategies has
physiological states have been attempted but have not yet paved the way for modeling other biological systems. For
been routinely adopted. While catalogs have been curated in example, attempts to map transcriptional gene regulatory
recent years describing the global regulation of mammalian networks have been pursued. Similar to metabolic networks,
genes, detailed biochemical and kinetic data associated with the first versions of these networks were constructed from the
enzymatic pathways controlling cellular metabolism and simple known regulatory pathways mapped via traditional
energy production have been available for decades. As a molecular biology studies. With the advent of high-
result, mathematical frameworks exist for quantifying how throughput expression array technology and the ability to
metabolic variables (eg, metabolite concentrations) depend quantify and correlate alterations in mRNA expression at the
on biochemical network parameters,2– 4 thus setting the stage transcriptomic level to various stimuli, the ability to construct
for attempts at utilizing in silico simulation and modeling to gene “coexpression networks” has advanced considerably,
understand complex biochemical processes. allowing for mapping of gene modules that are coregulated
Methodology involves use of either deterministic [eg, under a shared stimulus or are associated with a relevant
systems of ordinary differential equations (ODEs) to assess biological condition or genetic polymorphism (Figure 2A). In
the effects of perturbations on associated nodes in the this context, novel computational procedures have been
network context] or stochastic [eg, accounting for random developed, such as “expression screening,” which can inte-
effects on associated nodes by calculation of effect probabil- grate information from thousands of microarray expression
ities after network perturbation] simulation of reaction net- data sets to identify genes that are consistently coexpressed
works. Such simulation has allowed for the computation of with a target pathway, such as those in oxidative phosphoryla-
metabolite steady-state concentrations and their stability5 or tion,16 across biological contexts. By additionally accounting for
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
Chan and Loscalzo Network Medicine and Human Disease 361

Figure 1. An introduction to network-based


strategies in biomedical research. A, Hypotheti-
cal biological network. Essential genes (called
“hubs”) demonstrate dense interconnectivity with
other genes and hold a central topographical posi-
tion in the network; nonessential genes (called
“nodes”) tend to be placed at the network periph-
ery and are less well connected to other genes.
Genes that are responsible for normal biological
variability tend to be nonessential, whereas defi-
ciencies of “hub” genes typically cause embryonic
lethality.1 Many human disease genes (arrow) are
located at the periphery and display a preference
to interact with other disease genes (the “local
hypothesis”). Adapted from Chan et al,107 with per-
mission. B, The universe of “network medicine”
links genetic, “-OMIC,” biochemical, cellular, physi-
ological, and clinical data to create an intercon-
nected graph that can be used to model predic-
tively a (patho)biological event(s). Adapted from
Morel et al,108 with permission (illustration credit:
Ben Smith).

predictive algorithms to identify promoter binding sites for log.18 –22 To date, novel insight of biological function from
transcription factors (TRANSFAC and B-cell interactome) as these PPI networks has resulted largely from analysis of static
well as incorporating data from high-throughput assays demon- networks. There has been thought of establishing more
strating actual physical binding of transcription factors to pro- dynamic modeling via incorporation of reported association
moters [chromatin immunoprecipitation followed by microarray constants among protein pairs. However, it remains to be seen
analysis (ChIP-ChIP) and ChIP followed by sequencing (ChIP- if mathematical modeling can adequately capture the sheer
Seq) as found in databases including the Universal Protein complexity of these networks, and, more importantly, if such
Binding Microarray Resource for Oligonucleotide Binding Eval- modeling is accurate when considering the potential need to
uation (UniPROBE) and the open access database of transcrip- define the precise and changing concentrations of each
tion factor-binding profiles, JASPAR], further coexpression protein component involved in competitive binding to
networks have been verified and, in turn, used to identify novel multiple protein partners in different cellular compart-
factors and direct regulatory connections that influence substan- ments. Similarly, regulatory relationships that coordinate
tially various cellular phenotypes. activating and inhibitory posttranslational modifications
(eg, phosphorylation, acetylation, S-nitrosylation, redox
Network Modeling of Protein-Protein Interactions modifications, etc) have begun to be described in databases
and Posttranslational Modifications such as PhosphoELM, PhosphoSite, phosphorylation site
More recently, there have been considerable advances made database (PHOSIDA), NetPhorest, and the CBS prediction
in cataloguing the immense set of human protein-protein database. While phosphorylation sites in the proteome have been
interactions (PPI). While curation from the scientific litera- reasonably mapped and catalogued, the systematic cataloging of
ture alone has revealed substantial obstacles17 (Figure 2B), alternative modifications has yet to be completed. When con-
more promising methodologies using yeast 2-hybrid and sidering the more than 300 posttranslational modifications of the
3-hybrid screens as well as immunoprecipitation and high- proteome that are now recognized as well as associated protein
throughput mass spectrometry have greatly increased the binding complexes with bioactive inorganic ions (eg, iron,
likelihood of attaining a more comprehensive PPI cata- iron-sulfur clusters, selenium, etc), such network mapping re-
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
362 Circulation Research July 20, 2012

Figure 2. Network modeling of func-


tional interactions among biological
molecules. A, Topology of a gene coex-
pression module shared by developing
fetal myocardium and hypertrophied/fail-
ing myocardium. Adapted from Dewey et
al,53 with permission. B, Venn diagram
displaying the overlaps of the total
curated number of reported binary yeast
PPIs, with or without removing the larg-
est high-throughput yeast PPI reports
(numbers in parentheses). These data
suggest that that literature-curated data-
sets, at least for yeast, are far from com-
plete. Adapted from Cusick,17 with
permission.

mains an essential but daunting task. For instance, while sub- the systematic mapping of these modifications by mass spec-
stantial evidence supports the notion that redox modifications troscopy has begun by both academic23 and commercial biotech-
regulate specific enzymatic activities, identification and predic- nology laboratories.
tion of redox modifications throughout the proteome presents a
complicated challenge, as the exact identity of relevant modifi- Network Modeling of Epigenetic Modifications and
cations depends not only on protein site but also on the oxidative Interactions With Noncoding RNA
state of redox-sensitive amino acids, such as cysteines (eg, Epigenetic alterations of the genome (eg, DNA methylation)
sulfonic versus sulfenic versus thiolate moieties). Nonetheless, and the transcriptome (eg, repression by noncoding RNA)
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
Chan and Loscalzo Network Medicine and Human Disease 363

have recently been recognized as powerful and ubiquitous another.1 The resulting interactome displays significant clus-
gene regulatory mechanisms. These regulatory patterns are tering, with pockets of especially dense interconnectedness.
increasingly becoming incorporated into more comprehen- As a result, at most, a few links separate each node from any
sive gene regulatory networks. For example, the MethDB other node. Thus, perturbation of a single node can induce
database24 contains a catalog of genomic DNA methylation widespread effects throughout multiple modules and, thus,
encompassing 5382 methylation patterns or profiles for 48 can substantially alter multiple functional systems.
species, 1511 individuals, 198 tissues and cell lines, and 79 Importantly, the human interactome is a “scale-free” net-
phenotypes.25 Thus, it may soon be possible to integrate such work in which gene interactions do not occur randomly, with
granularity in this database into other networks to reveal the degree distribution following a power-law tail (Figure
novel functional connections through DNA methylation. 3A).1,35 Thus, this type of network contains a few highly
Numerous atlases also have been compiled recently detail- connected “hubs,” which are nodes that link substantially to
ing the systems-wide functions of noncoding RNA, such as multiple other nodes, and typically are essential for maintain-
microRNA (miRNA). MiRNA are conserved, non–protein- ing the integrity of the entire network. Hub proteins are
coding RNA molecules that have been identified as essential probably encoded by evolutionarily conserved genes that
mediators of a variety of genes and cellular processes. Inside function in crucial cellular activities. Some hubs appear to
the cell, miRNA negatively regulate gene expression primar- direct intrinsic gene cluster function, whereas others preserve
ily by binding to the 3! untranslated regions of messenger the integrity of the entire interactome by linking distinct
RNA (mRNA) transcripts to repress translation and/or de- clusters to one another. Scale-free networks exhibit properties
grade mRNA. Efficient binding relies on Watson-Crick that offer substantial insight into biological systems. Scale-
base-pairing between the seven-nucleotide “seed sequence” free properties minimize transition time between states of the
of a given miRNA and its mRNA target, and several system; they facilitate transfer of molecular information
algorithms (TargetScan, miRBase, PicTar, miRDB, and oth- across the system; they accommodate perturbations to the
ers) have accordingly been developed to predict accurately system with minimal adverse effects; and they promote
mRNA targets of each miRNA.26 In concert, high-throughput biological diversity.1 Notably, scale-free systems exhibit
analyses using transcriptional expression arrays and/or pro- “overdetermined” features, implying that the complete defi-
teomics have allowed for verification and identification of nition of all components of the network is not necessary to
additional miRNA targets,27 which have been incorporated understand how the network functions as a whole. Finally,
into a number of these databases (eg, DIANA and TarBase28). scale-free systems display emergent behavior, as defined by
Although the utility of these prediction algorithms and the fact that intimate knowledge of one node cannot be used
high-throughput methods has been reflected in their promi- to predict its behavior in a network nor the behavior of the
nent use in identifying key mRNA targets of specific miRNA, entire network (Figure 3B). It is this emergent property of
their use in more comprehensive network-based approaches biological networks that emphasizes the significant pitfalls in
is just beginning.29 –31 It is estimated that most messenger the reductionistic studies guiding biomedical research pres-
RNA transcripts are regulated by miRNA,32,33 with each ently—that is, erroneously attempting to extrapolate the
miRNA ("700 identified mammalian miRNA) typically functional study of a single gene alone to its in vivo role(s)
repressing expression of multiple mRNA (at times, "100 within the context of the endogenous biological network
targets simultaneously). It has been proposed that miRNA within which it operates.
appear to act by regulating multiple interacting targets in the Disease-relevant genes and related pathways are certainly
same functional network to generate robust actions in vivo.34 present within the human interactome but have been incom-
A deeper exploration of the implications of such networks of pletely defined. Currently, only 10% of human genes have
miRNA regulating networks of mRNA will certainly neces- been associated with a known disease (www.omim.org), and
sitate advances in computational analyses as well as experi- many have been found to correlate more often with nonhub
mental validation. Nonetheless, the possibility of mapping a nodes.36 Disease genes may also exist as hubs or essential
comprehensive epigenetic regulatory network for complex genes, but since dysfunction of essential genes often results in
human diseases is increasingly becoming realized. embryonic lethality, the association of disease genes and hubs
has been more difficult to prove, especially for chronic
Topological Characteristics of the diseases of adulthood. According to the “local hypothesis,”
Human Interactome disease genes also tend to interact directly with other disease
Building on the advances of mapping biochemical, gene genes in “disease modules” that induce a common pathophe-
regulatory, protein-protein, and epigenetic networks, we are notype36 (Figure 3C). Indeed, expansive analysis of gene
now able to construct a more expansive network that more expression arrays has confirmed the consistent and pervasive
comprehensively reflects the multitude of molecular connec- nature of these disease “signatures” of gene expression even
tions affecting human health and disease. Depending on across vastly different tissue sources.37 These disease mod-
which sets of databases are used, the human “interactome” ules can be challenging to map. Nonetheless, they may
currently encompasses #25 000 protein-coding genes, substantially overlap with “topological modules”—identifi-
#1000 metabolites, and an increasing number of posttrans- able by unbiased network-clustering tools—and with already-
lationally modified proteins and functional RNA molecules, defined “functional modules”— clusters of nodes of similar
together exceeding 100 000 participants and substantially or related function that have been defined previously by
greater functional interactions that variably overlap with one independent experimental validation38 (Figure 3C). Accord-
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
364 Circulation Research July 20, 2012

Figure 3. Properties of biological networks. A, The connectivity of random networks is reflected by a Poisson distribution, whereas a
scale-free (or clustered) network is defined by a power law distribution. As a result, there exist nodes that are sparsely linked as well as
nodes that are more highly linked (hubs). B, As depicted by a hypothetical complex electric circuit, the detailed analysis of 1 or 2 resis-
tors in isolation allows for little insight into the behavior of the circuit as a whole. Thus, similar to scale-free networks, this circuit dis-
plays emergent behavior. Moreover, detailed analysis of every resistor in the circuit is not necessary but rather can be inferred to
understand the circuit as a whole. Thus, scale-free networks and this circuit are overdetermined. C, Depiction of the 3 modularity con-
cepts of biological networks. Topological modules (left) are defined by locally dense neighborhoods of the interactome. Functional
modules (gray nodes, center) are defined by network neighborhoods in which there is a statistically significant segregation of nodes of
related function. Disease modules are groups of nodes whose perturbation (mutations, deletions, copy number variations, or expression
changes) can be linked to a particular disease phenotype and can cross functional molecules, shown as red nodes. Adapted from
Barabasi et al,1 with permission.

ingly, human disease manifestation may result from several called “neighborhood” or “clustering” methods) relies on
perturbations of a single disease module which may overlap network mapping of genes altered in transcriptional coexpres-
with the same components as related, but perhaps more easily sion analyses of diseased tissue, thus defining previously
identifiable, topological and functional modules.39 Conse- unrecognized factors carried within these “disease modules”
quently, the topographical position of a single disease gene in that influence pathogenesis. For cardiovascular diseases,
the human interactome can offer unique insight into its novel relevant modules have been defined for atherosclerosis,42– 45
associated partners, their connected roles in pathogenesis, and in-stent restenosis,45,46 type 2 diabetes mellitus,47 and cardiac
connected modules that regulate disease manifestation and hypertrophy and failure,48 among others. We have recently
response to therapeutic interventions. identified a disease module important in the progression of
pulmonary hypertension, allowing for the identification of a
Network Topology and Human Disease set of microRNA that control the pathophenotype based on
Network medicine can accurately predict novel disease genes, the predominance of their targets in that gene module29
based on their topographical position in relation to known (Figure 4B). Finally, delineation of modules that topolog-
disease-relevant factors (Figure 4A). First, the so-called ically neighbor but do not overlap with a given disease
“linkage” method enables the identification of unique factors gene or module has facilitated the delineation of new
important in human disease via their direct connection to disease pathways. Based on the mean topological distance
known disease genes. In cardiovascular disease, successful from a given set of disease nodes, so-called “random walk”
examples have included identification of a network of candi- algorithms have been useful in defining such neighboring
date susceptibility genes for coronary artery disease based on modules and linking them to diseases such as diabetes
disease genes selected from GWAS followed by analyses of mellitus.49 Linkage methods and mapping of disease mod-
directly connected neighboring genes via a protein-protein ules have become much more prevalent, probably due to the
interaction database.40 Ghazalpour et al identified a cadre of relative ease of performing these analyses. Conversely,
genes that are associated with obesity in the mouse based on diffusion methods appear to carry the best predictive
their links in a coexpression network with disease genes power,50 probably stemming from their ability to integrate
genetically associated with an obese phenotype.41 Second, the a greater amount of relevant topographical information
so-called “disease module” method (with some variants into the final analysis.
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
Chan and Loscalzo Network Medicine and Human Disease 365

Network medicine may also provide insight into the difficulty in inferring accurately initial or final concentrations
presumed but otherwise unappreciated molecular links of metabolites in a given condition. Second, the human
among separate human diseases. Goh et al first demonstrated network of metabolic reactions is more complex than that of
the existence of these widespread connections after construct- single-celled organisms. Finally, owing to the complexity of
ing a comprehensive map of disease modules in the human overlapping pathways and inputs in human tissue, rate con-
interactome36 (Figure 4C). Similarly, analyses of coexpres- stants are variable and are subject to alterations in enzyme
sion networks have revealed novel molecular connections activity, expression, and time-dependent relationships. None-
between Alzheimer disease and cardiovascular illnesses.51 theless, various mathematical models have been developed to
More recently, Rende et al constructed a cardiovascular reduce the impact of these constraints, leading to significant
disease functional linkage network (CFN) that carries signif- insight into the impact of metabolic dysfunction in human
icant interconnections among modules representing cardio- disease.
vascular diseases with other complex disorders such as Defining and applying the dynamic flux through gene
infection by Listeria monocytogenes, myasthenia gravis, regulatory networks presents an even greater challenge. Gene
hemorrhagic diatheses, and protein S deficiency.52 Gene coexpression networks alone falter as compared to more
coexpression analyses have also identified shared candidate sophisticated approaches to simulate reaction networks,
genes that link normal myocardial development to myocar- which stems from several causes. First, as generated from
dial hypertrophy and failure.53 Thus, such analyses can be expression array data, coexpression networks are relatively
used to identify links among developmental and pathological large and tend to encompass numerous subsets of molecular
contexts in the same tissue. As a result, these molecular networks. Coexpression alone is typically insufficient to map
interconnections may drive the clinical cosegregation of the direct regulatory connections among all relevant genes,
specific disease phenotypes. Furthermore, identification of and thus gene regulatory maps tend to remain incomplete.
these shared links may lead to novel, shared therapeutic Furthermore, unlike the previously described biochemical
targets for multiple diseases or suggest a “repurposed” use of networks in which each connection can typically be described
medications already approved for separate disease conditions. by a precise rate constant, connections functionally linking
gene regulatory networks together have not been as precisely
Network Dynamics and Human Disease defined, as binding affinity of transcription factors to gene
Although network topology certainly provides unique in- promoters do not necessarily always quantitatively reflect the
sights, more substantial advances in our comprehension of relative level of transcriptional activation. Finally, modeling
biological function in complex systems depend on under- dynamic changes in gene expression generally requires sto-
standing dynamic signal flux through these networks. To do chastic, rather than deterministic, approaches given the fre-
so, as with any complex circuitry, direct measurement or quently low concentrations of transcription factors and the
inference of three key components is essential: the initial limited number of promoter binding sites (as few as 2).
concentrations of each component or node, the overall struc- Despite these obstacles, methodologies have been estab-
ture of the network (eg, the interconnectivity), and the lished to infer better the exact nature of the relationships and
strength and direction of the nodal interactions. Although the rudimentary dynamics of gene regulatory networks. One
great advances have been made in defining the first two of method involves applied human genetics analysis. Presently,
these components in a variety of human disease conditions, chromosomal linkage mapping and GWAS are the most
accurate determination of the dynamically changing strength prevalent analytic methods used to reveal common and rare
of interactions is a challenging task. Nonetheless, rapidly genetic mutations and polymorphisms linked with human
advancing methodologies have been proposed to define or disease. These are powerful methods by which to identify
infer the details of those interconnections. novel genetic associations with human disease, but, when
As discussed previously, understanding the dynamics of used alone, these approaches are limited in their ability to
biochemical networks has been fruitful given the previously prove a causal or directed pathogenic mechanism. In contrast,
defined details regarding the network structure and the when coupled with an appreciation of where those genes
enzymatic rate constants of the reactions that interconnect lie in the interactome network, these genetic alterations can
within the network. Especially in the setting of the study of be integrated to infer the direction and dynamics of informa-
single-celled prokaryotes (eg, bacteria) or eukaryotes (eg, tion involving associated factors throughout the network to
yeast) where initial metabolite concentrations are readily determine causality among those molecular relationships. For
measured, precise calculations of sensitivity coefficients re- example, when used in an iterative and systematic process to
flect the predicted effects on the biochemical network after map how variations in DNA associate with relative transcript
specific perturbations. Application of such modeling has been abundances in a relevant coexpression network, statistical
powerful when performed in the context of bacterial patho- algorithms have been successful in inferring an independent,
genesis and has emphasized the importance of global meta- causative, or reactive function of disease-gene candidates
bolic alterations54 that contribute to bacterial growth, antibi- relative to a specific human disease phenotype.55 This ap-
otic susceptibility, and infective capabilities in humans. proach has been used for the identification of 3 new genes in
Modeling of metabolic networks in human tissue and human the susceptibility to human obesity and potentially could be
pathogenesis has been more challenging. First, complete applied to human mutations that segregate with overall
metabolomic profiles have only recently become available cardiovascular health risk (Figure 4D). More broadly, infer-
from clinically relevant tissue samples, thus increasing the ence of network dynamics through genetic study may help to
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
366 Circulation Research July 20, 2012

Figure 4. Discovery of novel disease genes using network-based approaches. A, Identification of novel disease genes using
network-based approaches. Adapted from Chan et al,107 with permission. B, A network-based strategy for identifying miRNA important
in pulmonary hypertension (PH) elucidates a target network of 7 miRNA groups at the crucial intersection of hypoxia, inflammation, and
TGF/BMP signaling. Such an approach also reveals genes that may represent points of coordinated miRNA regulation in PH. Circles
indicate predicted gene targets; triangles, miRNA; blue lines, predicted associations of miRNA and targets; dotted gray lines, gene
interactions documented in the consolidated interactome. Circle size is proportional to the number of miRNA groups (among these 7)
predicted to target that particular gene. Node colors represent functional pathways (ie, endothelin signaling, mitochondrial metabolism,
etc). Adapted from Parikh et al,29 with permission. C, A human “diseasome” displays molecular interconnections among separate cate-
gories of human disease. An edge denotes the involvement of a shared disease factor(s) or pathways common to multiple different dis-
eases. Adapted from Goh et al,36 with permission. D, Schadt et al used a likelihood-based causality model selection test (LCMS), using

Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014


Chan and Loscalzo Network Medicine and Human Disease 367

answer the question of “missing heritability” in human traits. Network Medicine and Drug Development
Specifically, despite efforts to identify additional genetic loci (Systems Pharmacology)
to explain the vastly differing patterns of heritability in In addition to aiding discovery of novel molecular aspects of
human disease, recent computational analyses by Zuk et al56 human disease, network medicine has the potential to revo-
suggest that variations in heritability need not invoke missing lutionize the process of drug discovery.64 Historically, drug
genetic variants but could adequately be explained by com- design has been based on key observations of pharmacolog-
plex variations in genetic interactions among different mo- ical or genetic manipulation of putative targets or pathways
lecular pathways. Thus, these findings emphasize the intrigu- that, when perturbed, alter a quantifiable (patho)phenotype.
ing potential of studying network dynamics to reveal a Unwanted pharmacological side effects have been attributed
widespread “network-based” origin of disease heritability. to an interaction of a medication with alternative factors in
Separate from human genetic studies, an alternative meth- separate pathways. However, in complex human diseases,
ods by which to study network dynamics in human disease both therapeutic and adverse consequences of drug treatment
involves “reverse engineering” regulatory networks.57 In this may depend on shared effectors and pathways that, when
approach, a discrete and typically small module of relevant manipulated in distinct combinations involving specific cel-
genes is systematically perturbed with measurement of the lular or tissue contexts, may drive disparate pathophenotypes.
consequent changes of their associated genes. The regulatory It has been estimated that approximately 400 to 600 proteins
relationships among the genes can then be inferred, yielding of the presumed 100 000 participants in the consolidated
novel predictions of direct regulatory connections that can be interactome are targets of the 1200 Food and Drug Admin-
verified experimentally. Indeed, such processes have been istration (FDA)-approved drugs65; however, a large propor-
validated in small sets of discrete gene networks active in tion of these established drugs do not target actual disease-
organisms, such as bacteria,58 yeast,59 and Drosophila,60 as
associated proteins but merely neighboring proteins.66 Taken
well as for mapping networks of small noncoding RNA in
together, these drugs may only modestly affect disease
bacteria.61 This approach has recently been applied to human
phenotype. In addition, there likely exist a larger number of
disease by Ergun et al, who inferred a gene regulatory
potential therapeutic targets that await identification.
network important in prostate cancer that led to validation of
Furthermore, despite the rapid advances in our knowledge
the androgen receptor, in the context of this network, as a
regarding complex biological processes, pharmaceutical re-
marker to detect relatively aggressive primary prostate can-
search and development productivity has been in decline
cers (Figure 4E).62 The advantage of such reverse engineering
since the mid-1990s (Figure 5A). In striking statistics drawn
is its unbiased approach to network mapping,63 as a precon-
from the pharmaceutical industry (2000 to 2008), only #25 to
ceived framework of regulatory connections is not necessary;
however, this approach is hampered by the technical chal- 30 new molecular entities per year are identified as viable
lenges of applying the algorithms to larger and much more therapeutic agents. Clinical development of these compounds
complicated regulatory networks. Additionally, because in- necessitates an average of 13.9 years, and new agents carry a
ferred relationships can only be depicted dichotomously as probability of success of approximately 2.01% per year.67
direct “activating” or “inhibiting” conditions, more complex Reasons for such declining productivity include a more
network relationships that involve connections beyond tran- intense regulatory environment, the exhaustion of “easy” or
scriptional regulation (ie, posttranslational modifications, more obvious targets, an increasing attrition rate for devel-
protein-protein oligomerization, etc) require different types of oping drugs secondary to their unanticipated side effects, and,
representation, and thus the accuracy of any simulation of from the perspective of this review, an intrinsically flawed
network perturbation could suffer. Future iterations of anal- reductionistic approach to drug development centered on the
ysis of the dynamics of gene regulatory networks may benefit perceived need to identify a single drug target that entirely
from utilizing the known topology of the existing consoli- reverses a given disease phenotype (ie, an Erlichian “magic
dated interactome coupled with statistical algorithms that bullet”). Conversely, the failures of single-target drugs in
incorporate the results of perturbations of genetic coexpres- treating complex human disease have already suggested their
sion networks via genetic associations and/or alternative severe limitations when used alone and have prompted the
stimuli and thus iteratively annotate and optimize the regu- exploration of potent combinatorial drug therapies for a
latory map. Consequently, a more sophisticated and compre- variety of disease phenotypes (ie, tuberculosis, cancer, HIV/
hensive network representation is possible that could be AIDS, heart failure, etc). Furthermore, given the advances in
applied to further systems-level discoveries of how patho- pharmacogenomics and improved understanding of how
genic or therapeutic stimuli may alter overall disease drugs may be differentially effective based on individual
manifestation. genotype, development of optimal drug regimens based on

Figure 4 (Continued). quantitative trait loci (QTL) data, to infer relationships between RNA levels and complex traits that are best sup-
ported by the data. Possible relationships (M1-M5) are shown here among QTL (L), RNA levels of a gene (R), and a complex trait (C).
Adapted from Schadt et al,55 with permission. E, Strategy of reverse engineering to infer a gene regulatory network in nonrecurrent pri-
mary and metastatic prostate cancer. In phase 1, high-throughput microarray data obtained from cancer cell lines or patient samples
are incorporated by the MNI algorithm to infer a model of regulatory interactions between genes (blue-filled circles indicate genes;
arrows indicate regulatory effects). In phase 2, test condition expression data are filtered using the reconstructed network to distin-
guish the genes affected by a condition (red-filled circles) from the remaining genes that exhibit changes in expression. Adapted from
Ergun et al,62 with permission.

Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014


368 Circulation Research July 20, 2012

Figure 5. Rationale for systems pharmacology. A, The declining productivity of pharmaceutical research is reflected by the steep
decline in the number of newly approved medications over the past decade. B, Comparison of the models used in conventional and
systems pharmacology to study the effects of drugs on a given system. C, Proposed approach for network-based drug discovery
involving first defining a disease module and then iteratively refining the map based on its emergent properties to identify appropriate
drug targets. Adapted from Loscalzo64 and Schadt et al,109 with permission.

their individualized effects is under further investigation and Identification and Targeting Novel
of premium priority.68,69 Drug-Host Interactions
Rapidly evolving methods of “systems pharmacology”70 Application of systems pharmacology to the identification of
facilitate a more rational and efficient approach to identifying novel drug-host interactions in complex human disease has
the most relevant “network-based” targets based on their been reported with success. To identify a larger pool of
presence in relevant disease modules (Figure 5B).64 Given the potential therapeutic targets in human disease that are not
expansive small molecule libraries that have been mapped to limited by preexisting knowledge of disease genes, systems
direct targets through large scale chemical biology reposito- pharmacology has been employed to generate a quantitative
ries,71 network theory presents the opportunity not only to framework within which to compare and contrast diseases by
discern previously unidentified targets for disease therapy an integrated analysis of disease-related mRNA expression
and to map novel pathogenic pathways but also to predict data and the human protein interaction network. For example,
how those targets can be manipulated, in isolation or in Lamb et al have devised a process (“Connectivity Map”) of
combination. A generalized methodology has recently been identifying drug-dependent gene expression profiles that
proposed for a systems-based method for drug discovery that correlate with known disease-dependent profiles,73 thus al-
involves defining a disease module and iteratively refining lowing for a method for in silico screening of drugs that may
the map based on its emergent properties (Figure 5C).64 In have salubrious effects in multiple disease contexts. A variant
doing so, drugs can be predicted and/or produced that of that approach has been pursued by Hu and Agarwal,74 in
rationally target key disease nodes followed by iterative which gene expression data are not prospectively collected
refinement in silico that account for its effects on the but can be directly obtained from an existing database, Gene
emergent behavior of the disease module. Therefore, contrary Expression Omnibus.75 A “guilt by association” method has
to traditional drug design, systems pharmacology may offer also been proposed by Chiang and Butte,76 whereby predic-
elegant and cost-effective solutions to identifying multiple tions of novel associations between drugs and diseases can be
drug combinations (“rational polypharmacy”)64 that maxi- made by assuming that if two diseases are treated by the same
mize the therapeutic efficacy, minimize adverse side effects, drug, alternative drugs treating only one of them might also
and minimize drug-drug interactions. As a result, we may treat the other. More recent studies have attempted to make
expect a much lower attrition rate as these agents are predictions on a broader scale. Suthram et al identified 4620
clinically evaluated, decreased time from drug conception to functional modules in the human protein network and pro-
approval, and substantially higher rate of success for all vided a quantitative metric to record their responses in 54
compounds that enter clinical testing (provided regulatory human diseases leading to 138 significant similarities be-
bodies can adapt to this change in drug development strategy tween diseases77 (Figure 6A). Interestingly, 14 of the signif-
and clinical trial design strategies [adaptive] follow suit72). icant disease correlations also shared common drugs, support-
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
Chan and Loscalzo Network Medicine and Human Disease 369

Figure 6. Current examples of drug discovery by systems pharmacology. A, Network of 138 significant disease correlations, 14 of
which encompass shared drug targets. The colors correspond to significant disease correlation categories after hierarchical clustering.
Adapted from Suthram et al,77 with permission. B, Reverse-engineered pathways related to aminoglycoside lethality. On comparing
treatment with a lethal aminoglycoside versus the bacteriostatic ribosome inhibitor, spectinomycin, gene expression changes were fil-
tered through an E coli gene connectivity map; the resulting gene networks were analyzed for functional enrichment. The strains exhib-
iting decreased growth or increased growth found from a high-throughput screen of an E coli single-gene deletion library treated with
gentamicin were overlaid on these networks. Adapted from Kohanski et al,87 with permission. C, Network map displaying the relation of
LQTS seed nodes (red) with 2 distinct drugs (blue), the Src inhibitor dasatinib (a cancer drug), and loperamide (an antidiarrheal) that
interacts with calmodulin (CALM1). Adapted from Berger et al,99 with permission. D, Differential side effects of CETP inhibitors (torce-
trapib, anacetrapib, and JTT705 or dalcetrapib) are predicted by their complex interactions with the PPAR network. Adapted from Xie
et al,101 with permission.

ing the hypothesis that similar diseases can be treated by the side effects, etc), networks of drug targets, and clinical
same drugs, allowing for predictions for new uses of existing pathophenotypic similarities, a more comprehensive compen-
drugs. Finally, the authors identified 59 modules that are dium can be developed of all relevant drug indications for
significantly enriched for genes that are known to be drug human disease.78 Consequently, these types of studies may
targets and are dysregulated in at least half of the studied form the computational foundation for future development of
diseases, representing a common disease-state “signature” personalized therapies, in which variations in gene expression
and highlighting the importance of these core modules as signatures and specific phenotypes from individual patients
prime therapeutic opportunities. Intriguingly, Gottlieb et al could be considered in order to dictate the course of care.
have described a novel method (PREDICT) for the large- Advances in “repurposing” or “repositioning” existing
scale prediction of drug indications. This approach is predi- drugs for alternative diseases have also benefited from
cated on the idea that similar drugs are indicated for similar network methodology. High-throughput methods of literature
diseases. Thus, by quantifying and considering the known curation have proven effective in defining novel drug-disease
similarities among drugs (ie, chemical structure, predicted connections that can guide repurposing of known medica-
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
370 Circulation Research July 20, 2012

tions.79 For example, Li et al have constructed disease- further perturbed these bacterial networks leading to mis-
specific (eg, Alzheimer disease, in this case) drug-protein translation and misfolding of membrane proteins central to
connectivity maps derived solely from literature database aminoglycoside-induced oxidative stress and cell death, thus
searches as a way to uncover a more comprehensive set of expanding our understanding of the common mechanism of
known drugs acting in the same disease context.80 More killing induced by known bactericidal antibiotics (Figure
recently, commercial bioinformatics companies specializing 6B).87 Furthermore, potent new antimicrobial agents have
in the analysis of scientific literature, such as Ingenuity, been identified and validated de novo based on their predicted
GeneGo, and Adriadne Genomics, have offered proprietary system-based metabolic responses on bacterial survival.54
pathway- and network-based solutions to the pharmaceutical Our understanding of systems-wide metabolic alterations
industry for drug repurposing.81 Beyond literature curation, in diseased mammalian cells has also led to novel potential
other groups have further developed a systematic computa- therapeutic targets. For instance, by mapping the dynamic
tional approach to predict novel therapeutic indications on the fluxes of labeled metabolites in mammalian cells, Munger et
basis of comprehensive testing of molecular signatures in al noted an especially prominent increase in fatty acid
drug-disease pairs.82 For instance, Sirota et al integrated gene biosynthesis during human cytomegalovirus (HCMV) infec-
expression measurements from 100 diseases and gene expres- tion.88 Subsequent pharmacological targeting of fatty acid
sion measurements on 164 drug compounds, yielding pre- biosynthesis suppressed the replication of HCMV and influ-
dicted therapeutic potentials for these drugs. Previously enza A, thereby confirming that fatty acid synthesis is
validated drug and disease relationships were predicted, as essential for the replication of two divergent viruses and,
well as a number of novel indications for these 164 drugs. importantly, that systems-level metabolic flux profiling can
Importantly, one such prediction was experimentally vali- identify metabolic targets for antiviral therapy.
dated for the antiulcer drug cimetidine as a candidate thera- In the field of cancer biology, Otto Warburg first described
peutic in the treatment of lung adenocarcinoma, demonstrat- a half a century ago that tumors exhibit enhanced glycolysis
ing its efficacy in vivo using mouse xenograft models.83 In a coupled with a reduction in oxidative phosphorylation, de-
separate set of experiments, another prediction regarding the spite adequate oxygen availability. This “Warburg effect”
potential for the anticonvulsant topiramate in treatment of was proposed to be a key driver of tumor progression.89 Since
inflammatory bowel disease was successfully validated in a that initial observation, we now recognize that there exists a
rodent model of inflammatory bowel disease.84 As a result, systems-wide “reprogramming” of metabolic pathways asso-
the use of existing gene coexpression networks in combina- ciated with alterations in bioenergetics and mitochondrial
tion with the known molecular interactions of existing drugs function in transformed cells; for this reason, metabolic
has yielded more comprehensive insight into the pleiotropic interventions are now emerging as potential therapeutic
effects of various medicinal compounds. antineoplastic strategies.90 –92 Accordingly, the fine detail of
A variation on the theme of repurposing drugs includes the systems-level profiling available in cancer biology93,94 has
notion of rationally “rewiring” molecular networks such that allowed for a growing number of network-based studies
cells resistant to certain medications can become responsive reporting novel metabolic pathways important in driving
to pharmacological exposure. Recently, Lee et al undertook cellular transformation and thus identifying crucial metabolic
such a project, utilizing an expansive systems-level approach targets for pharmacological repression.95,96 Conversely, in the
using comprehensive time-dependent measurements of sig- future, pharmacological rescue of metabolic dysfunction
naling networks, gene expression profiles, and cell pheno- could be attempted in genetic metabolic diseases as a novel
typic responses in combination with computational network alternative to gene therapy.
modeling of breast cancer cells resistant to genotoxic drugs.85
In doing so, the authors constructed and validated a molecular Identification of “Off-Target” Effects and
roadmap to reprogram sequentially oncogenic signaling path- Drug-Drug Interactions
ways and resensitize breast cancer cells to those drugs. Such In addition to guiding the rational targeting of specific
results indicate the feasibility of not only studying, but more pathways in complex diseases, network-based approaches
importantly manipulating, large-scale network dynamics to- have better informed us about the systems properties of a full
ward a desired cellular phenotype. drug interaction network that encompass currently unwanted
Perhaps the most striking successes of network pharmacol- but otherwise unanticipated (so-called “off-target”) effects
ogy to date are derived from studies of bacterial metabolism. and drug-drug interactions. For example, Yeh et al created a
Owing to the fine detail of both the topology and rate network map of drug-drug interactions using a unique biolu-
constants connecting metabolic networks in the bacterial minescence technique to provide quantitative measurements
interactome, in silico predictions have been possible regard- of pairwise interactions among 21 antibiotics that affect
ing the effects on metabolite flux and consequent phenotypic growth rate in Escherichia coli.97 Interestingly, the investi-
alterations after perturbation of specific nodes in those gators reported that the drug interaction network revealed 2
pathways. As a result, by combining phenotypic and genetic classes of drug that interact either purely synergistically or
experiments with microarray analyses to identify relevant purely antagonistically—network properties that correspond
bacterial networks important in antibiotic response, Kohanski directly to the cellular functions affected by the drugs. This
et al have demonstrated that all classes of bactericidal network approach introduced a new conceptual framework
antibiotics, regardless of their specific target, promote the for understanding the functional mechanisms of drugs and
generation of lethal hydroxyl radicals.86 Kohanski et al have their cellular targets, which could be expanded to other
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
Chan and Loscalzo Network Medicine and Human Disease 371

FDA-approved medications or small molecule libraries. In- hypertension (Figure 6D). Furthermore, using these networks
terestingly, Torella et al have extended these studies of as a guide, the adverse drug effects of second generation
antibiotic synergy by utilizing mathematical modeling of the CETP inhibitors could be minimized by fine-tuning multiple
in vivo dynamics of bacterial infection to determine the off-target interactions using single or multiple therapies.
optimal strategies for utilizing synergism and antagonism to Similarly, network approaches may also be effective in
suppress better the evolution of multi-drug resistance.98 predicting drug-drug interactions. Hansen et al have de-
Analysis of the consolidated interactome can yield even scribed their preliminary experience with studying the antidi-
greater insight into these pleiotropic and complicated molec- abetic thiazolidinedione, rosiglitazone, which has been with-
ular interactions among various drugs. Recently, such a drawn from the market due to serious adverse side effects.65
pharmacological approach was applied to the long-QT syn- According to the FDA-AERS, rosiglitazone is associated with
drome (LQTS) to determine whether a network-based anal- significant thrombotic complications as well as peripheral
ysis could predict arrhythmic side effects of known FDA- edema and total body volume overload. Consequently, these
approved medications (Figure 6C).99 It is already established authors have constructed protein-protein interaction networks
that mutations in any of 12 genes encoding ion channels are based on Gene Ontology terms that relate to thrombosis and
linked with congenital LQTS and potentially fatal ventricular fluid regulation. To determine the degree of overlap, these
arrhythmias, such as torsades de pointes. In an elegant study, subnetworks were then cross-referenced with direct gene
Berger et al reasoned that drugs targeting proteins located in targets of PPAR!, a well-established direct target of rosigli-
the network “neighborhood” of bona fide LQTS genes should tazone. Via utilization of FDA-AERS, drugs were then
also reveal arrhythmic side effects. On construction of a identified, which, when used in combination with rosiglita-
protein-protein interactome, a LQTS “neighborhood” of zone, should increase or decrease associated complications of
genes was identified by use of a mean first passage time thrombosis or edema by analyzing alterations in the effectors
algorithm to reveal proteins that interact directly or with close shared as targets of rosiglitazone and as members in the above
neighbors of seed LQTS genes. Importantly, the known subnetworks. These authors continue to develop algorithms to
targets of certain FDA-approved drugs that are associated predict whether combinations of drugs may target the same
with LQTS as an adverse event were enriched in the LQTS
pathway and whether they could potentiate therapeutic or
neighborhood. By screening the remaining cache of FDA-
adverse consequences.
approved medications for those that target the LQTS neigh-
borhood, additional drugs were identified that probably in-
Conclusions
fluence QT length. These predictions were then validated by
Network medicine offers insight into the integrative nature of
cross-referencing with the Adverse Events Reporting System
human pathogenesis by considering the dense interconnec-
of the FDA (FDA-AERS). Thus, identification and utilization
tions among disease genes rather than merely considering
of disease-selective neighborhoods within the human interac-
those genes in isolation. It allows for novel predictions of
tome can provide specific insight into predicting adverse
disease genes, the interconnected actions of those genes in
event susceptibility for new and “repurposed” drugs. Re-
pathogenesis, the molecular relationships among separate
cently, Tatonetti et al have developed comprehensive,
disease conditions, and the concerted response to therapeutic
network-driven databases of drug effects and of drug-drug
interaction side effects and demonstrated novel predicted interventions. Although still in its early stages, this evolving
adverse effects among cardiovascular drug classes.100 discipline offers a potentially powerful set of concepts and
Alternatively, network approaches offer substantial insight tangible methods by which to improve rapidly our molecular
into the elucidation of the unanticipated “off-target” effects of understanding of the networked nature of human disease and
otherwise presumed direct targeted therapies. Pharmacologi- to design medical therapies rooted in the complexity of these
cal examples of this scenario are plentiful, such as the biological connections. Nonetheless, despite benefiting from
well-documented clinical failure of the cholesteryl ester the overdetermined property of biological networks, the
transfer protein (CETP) inhibitor torcetrapib.101 Despite its fidelity of network medicine is limited by the currently
efficacy in augmenting HDL levels and reducing triglyceride incomplete nature of the interactome maps. Yet, while many
levels, the use of torcetrapib surprisingly incurred risk of predictions derived from network methodology await confir-
substantial hypertension, prompting a halt to its development mation, specific fundamental network principles have been
following phase III clinical trials.102,103 Based on in silico consistently supported by independent experimental proof in
predictions of protein-ligand binding, Xie et al identified a human and animal models of disease. Such methods are even
panel of off-targets for torcetrapib and other CETP inhibitors beginning to permeate the commercial biotechnology sector,
from the human structural genome and mapped those targets as exemplified by companies such as Proteostasis (www.
to a network of biological pathways curated from the litera- proteostasis.com), which was founded on the concept of
ture.101 The predicted protein-ligand network was consistent using systems pharmacology to target efficiently the “protea-
with experimental results revealing that the side effect of some” gene network. In the very near future, we expect a
hypertension by torcetrapib could be predicted by its target- more widespread expansion of network methodology in
ing of the peroxisome proliferator-activated receptor (PPAR) biomedicine. Consequently, “systems pathophysiology” may
system. Importantly, second-generation CETP inhibitors are become more of a reality as we explore not only intracellular
not predicted to have the same magnitude of action on the molecular networks but also the even less well understood
PPAR system and thus a lower likelihood of inducing spatio-temporal interconnections among distinct cells, tis-
Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014
372 Circulation Research July 20, 2012

reclassify human disease and thus allow for a more expansive


consideration of the pathogenic mechanisms that are truly at
play (Figure 7).106 By incorporating the relationships of
individual disease genes, their connections within a disease
module, and that module’s shared links with separate disease
modules, insight can be garnered regarding the coordinated
actions of several disease modules as well as specific molec-
ular links that represent “intermediate” phenotypes (endop-
athophenotypes) shared among various disease conditions (ie,
inflammation, thrombosis, etc). Furthermore, genetic, envi-
ronmental, and even behavioral “nodes” can be represented
accurately by this approach. As a result, this type of disease
Figure 7. Network-based classification of human disease. In classification could predict a highly individualized clinical
this model, a specific disease module (white nodes), associated pathophenotype that ultimately represents a more accurate
with myocardial ischemia (module 1) and containing a known
disease gene (arrow), is functionally linked to 2 disease mod-
molecular schema on which to base future discovery in
ules (modules 2 and 3; gray nodes) by shared nodes (black) and personalized medicine.
edges. These shared nodes may regulate intermediate (patho)
phenotypes important in other disease processes, such as Acknowledgments
inflammation or thrombosis. Influenced by a combination of
environmental and genetic events that also regulate these dis- We thank Stephanie Tribuna for expert administrative assistance and
ease networks, individualized manifestations of myocardial Sol Chan for assistance with figures.
dysfunction result (early myocardial infarction, ventricular
tachycardia, or ischemic cardiomyopathy). By incorporating an Sources of Funding
understanding of network function in these biological circuits, This work was supported in part by the NIH, the Pulmonary
these manifestations may be better predicted, treated, and pre- Hypertension Association, Gilead Sciences, and the Lerner, Harris,
vented. Adapted from Chan et al,107 with permission.
and Watkins funds (S.Y.C.); and NIH grants HL061795, HL48743,
HL107192, HL70819, and HL108630 (J.L.).
sues, and organ systems that have just begun to be explored
in cardiac morphogenesis.104 Disclosures
Thus far, network medicine has benefited from the exper- None.
tise of computer scientists, engineers, statistical physicists,
theoretical biologists, and biostatisticians to generate a range References
of predictions regarding the systems-wide behavior of com- 1. Barabasi AL, Gulbahce N, Loscalzo J. Network medicine: a network-based
approach to human disease. Nat Rev Genet. 2011;12:56–68.
plex biological systems that otherwise had not been accessi-
2. Salter M, Knowles RG, Pogson CI. Metabolic control. Essays Biochem.
ble. Such work has led to the recent establishment of a 1994;28:1–12.
number of highly regarded journals that specialize in compu- 3. Heinrich R, Schuster S. The Regulation of Cellular Systems. New York,
tational biological approaches, such as PLoS Computational NY: Chapman and Hall; 1996.
4. Fell D. Understanding the Control of Metabolism. London, UK:
Biology, BMC Bioinformatics, and Molecular Systems Biol-
Portland Press; 1997.
ogy, among others. However, most contemporary reports of 5. Schuster S, Heinrich R. The definitions of metabolic control analysis
applying network theory to biomedical research have resulted revisited. Biosystems. 1992;27:1–15.
in predictions that remain theoretical and are yet to be 6. Friedland AE, Lu TK, Wang X, Shi D, Church G, Collins JJ. Synthetic
validated. Most likely, this shortcoming stems from limita- gene networks that count. Science. 2009;324:1199 –1202.
7. Perumal TM, Gunawan R. Understanding dynamics using sensitivity
tions in effective communication among computational sci- analysis: caveat and solution. BMC Syst Biol. 2011;5:41.
entists, molecular and biomedical scientists, and translational 8. Alves R, Antunes F, Salvador A. Tools for kinetic modeling of bio-
physician-scientists. Thus, theories are published that are not chemical networks. Nat Biotechnol. 2006;24:667– 672.
perceived as relevant to biomedical or translational science, 9. Chen WW, Niepel M, Sorger PK. Classic and contemporary approaches
to modeling biochemical reactions. Genes Dev. 2010;24:1861–1875.
are not easily validated using experimental biology, and, 10. Ihekwaba AE, Broomhead DS, Grimley RL, Benson N, Kell DB. Sen-
perhaps most importantly, are not described in terms that are sitivity analysis of parameters controlling oscillatory signalling in the
easily accessible to colleagues with vastly different scientific NF-kappaB pathway: the roles of IKK and IkappaBalpha. Syst Biol.
backgrounds. A more facile ability to integrate these currently 2004;1:93–103.
11. Ihekwaba AE, Broomhead DS, Grimley R, Benson N, White MR, Kell
diverse scientific fields is warranted, and efforts are under- DB. Synergistic control of oscillations in the NF-kappaB signalling
way to train more formally the next generation of biologists pathway. Syst Biol. 2005;152:153–160.
and biomedical scientists in network and systems ap- 12. Chu Y, Jayaraman A, Hahn J. Parameter sensitivity analysis of IL-6
proaches.105 In the meantime, the great potential of network signalling pathways. IET Syst Biol. 2007;1:342–352.
13. Iwamoto K, Tashima Y, Hamada H, Eguchi Y, Okamoto M. Mathe-
biology in translational medicine fundamentally relies on the matical modeling and sensitivity analysis of G1/S phase in the cell cycle
close collaboration of diverse scientists who can integrate including the DNA-damage signal transduction pathway. Biosystems.
their expertise to coordinate the vast information contained in 2008;94:109 –117.
biological networks into hypotheses that are testable by the 14. Adler P, Peterson H, Agius P, Reimand J, Vilo J. Ranking genes by their
co-expression to subsets of pathway members. Ann N Y Acad Sci.
experimentalist. 2009;1158:1–13.
Finally, by integrating the dense interconnections among 15. Mardinoglu A, Nielsen J. Systems medicine and metabolic modelling.
disease networks, a new framework has been proposed to J Intern Med. 2012;271:142–154.

Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014


Chan and Loscalzo Network Medicine and Human Disease 373

16. Baughman JM, Nilsson R, Gohil VM, Arlow DH, Gauhar Z, Mootha 40. Jensen MK, Pers TH, Dworzynski P, Girman CJ, Brunak S, Rimm EB.
VK. A computational screen for regulators of oxidative phosphorylation Protein interaction-based genome-wide analysis of incident coronary
implicates SLIRP in mitochondrial RNA homeostasis. PLoS Genet. heart disease. Circ Cardiovasc Genet. 2011;4:549 –556.
2009;5:e1000590. 41. Ghazalpour A, Doss S, Zhang B, Wang S, Plaisier C, Castellanos R,
17. Cusick ME, Yu H, Smolyar A, Venkatesan K, Carvunis AR, Simonis N, Brozell A, Schadt EE, Drake TA, Lusis AJ, Horvath S. Integrating
Rual JF, Borick H, Braun P, Dreze M, Vandenhaute J, Galli M, Yazaki genetic and network analysis to characterize genes related to mouse
J, Hill DE, Ecker JR, Roth FP, Vidal M. Literature-curated protein weight. PLoS Genet. 2006;2:e130.
interaction datasets. Nat Methods. 2009;6:39 – 46. 42. Wang SS, Schadt EE, Wang H, Wang X, Ingram-Drake L, Shi W, Drake
18. Rual JF, Venkatesan K, Hao T, et al. Towards a proteome-scale map of TA, Lusis AJ. Identification of pathways for atherosclerosis in mice:
the human protein-protein interaction network. Nature. 2005;437: integration of quantitative trait locus analysis and global gene expression
1173–1178. data. Circ Res. 2007;101:e11– e30.
19. Stelzl U, Worm U, Lalowski M, et al. A human protein-protein inter- 43. Diez D, Wheelock AM, Goto S, Haeggstrom JZ, Paulsson-Berne G,
action network: a resource for annotating the proteome. Cell. 2005;122: Hansson GK, Hedin U, Gabrielsen A, Wheelock CE. The use of network
957–968. analyses for elucidating mechanisms in cardiovascular disease. Mol
20. Ewing RM, Chu P, Elisma F, et al. Large-scale mapping of human Biosyst. 2010;6:289 –304.
protein-protein interactions by mass spectrometry. Mol Syst Biol. 44. Romanoski CE, Che N, Yin F, Mai N, Pouldar D, Civelek M, Pan C, Lee
2007;3:89. S, Vakili L, Yang WP, Kayne P, Mungrue IN, Araujo JA, Berliner JA,
21. Venkatesan K, Rual JF, Vazquez A, et al. An empirical framework for Lusis AJ. Network for activation of human endothelial cells by oxidized
binary interactome mapping. Nat Methods. 2009;6:83–90. phospholipids: a critical role of heme oxygenase 1. Circ Res. 2011;109:
22. Dreze M, Monachello D, Lurin C, Cusick ME, Hill DE, Vidal M, Braun e27– e41.
P. High-quality binary interactome mapping. Methods Enzymol. 2010; 45. Civelek M, Manduchi E, Riley RJ, Stoeckert CJ Jr, Davies PF. Coronary
470:281–315. artery endothelial transcriptome in vivo: identification of endoplasmic
23. Zhao C, Sethuraman M, Clavreul N, Kaur P, Cohen RA, O’Connor PB. reticulum stress and enhanced reactive oxygen species by gene connec-
Detailed map of oxidative post-translational modifications of human tivity network analysis. Circ Cardiovasc Genet. 2011;4:243–252.
p21ras using Fourier transform mass spectrometry. Anal Chem. 2006; 46. Ashley EA, Ferrara R, King JY, et al. Network analysis of human
78:5134 –5142. in-stent restenosis. Circulation. 2006;114:2644 –2654.
24. Grunau C, Renault E, Rosenthal A, Roizes G. MethDB: a public 47. Sengupta U, Ukil S, Dimitrova N, Agrawal S. Expression-based network
database for DNA methylation data. Nucleic Acids Res. 2001;29: biology identifies alteration in key regulatory pathways of type 2
270 –274. diabetes and associated risk/complications. PLoS One. 2009;4:e8100.
25. Negre V, Grunau C. The MethDB DAS server: adding an epigenetic 48. Dewey FE, Wheeler MT, Ashley EA. Systems biology of heart failure,
challenges and hopes. Curr Opin Cardiol. 2011;26:314 –321.
information layer to the human genome. Epigenetics. 2006;1:101–105.
49. Vanunu O, Magger O, Ruppin E, Shlomi T, Sharan R. Associating genes
26. Bartel DP. MicroRNAs: target recognition and regulatory functions.
and protein complexes with disease via network propagation. PLoS
Cell. 2009;136:215–233.
Comput Biol. 2010;6:e1000641.
27. Watanabe Y, Kanai A. Systems biology reveals microRNA-mediated
50. Navlakha S, Kingsford C. The power of protein interaction networks for
gene regulation. Front Genet. 2009;2:29.
associating genes with diseases. Bioinformatics. 2010;26:1057–1063.
28. Vergoulis T, Vlachos IS, Alexiou P, Georgakilas G, Maragkakis M,
51. Ray M, Ruan J, Zhang W. Variations in the transcriptome of Alzhei-
Reczko M, Gerangelos S, Koziris N, Dalamagas T, Hatzigeorgiou AG.
mer’s disease reveal molecular networks involved in cardiovascular
TarBase 6.0: capturing the exponential growth of miRNA targets with
diseases. Genome Biol. 2008;9:R148.
experimental support. Nucleic Acids Res. 2011;40(Database issue):
52. Rende D, Baysal N, Kirdar B. A novel integrative network approach to
D222–D229.
understand the interplay between cardiovascular disease and other
29. Parikh VN, Jin RC, Rabello S, et al. MicroRNA-21 integrates
complex disorders. Mol Biosyst. 2011;7:2205–2219.
pathogenic signaling to control pulmonary hypertension: results of a
53. Dewey FE, Perez MV, Wheeler MT, Watt C, Spin J, Langfelder P,
network bioinformatics approach. Circulation. 2012;125:1520 –1532. Horvath S, Hannenhalli S, Cappola TP, Ashley EA. Gene coexpression
30. Lin CC, Chen YJ, Chen CY, Oyang YJ, Juan HF, Huang HC. Crosstalk network topology of cardiac development, hypertrophy, and failure. Circ
between transcription factors and microRNAs in human protein inter- Cardiovasc Genet. 2010;4:26 –35.
action network. BMC Syst Biol. 2012;6:18. 54. Shen Y, Liu J, Estiu G, Isin B, Ahn YY, Lee DS, Barabasi AL, Kapatral
31. Gennarino VA, D’Angelo G, Dharmalingam G, Fernandez S, Russolillo V, Wiest O, Oltvai ZN. Blueprint for antimicrobial hit discovery tar-
G, Sanges R, Mutarelli M, Belcastro V, Ballabio A, Verde P, Sardiello geting metabolic networks. Proc Natl Acad Sci U S A. 2010;107:
M, Banfi S. Identification of microRNA-regulated gene networks by 1082–1087.
expression analysis of target genes. Genome Res. 2012;22:1163–1172. 55. Schadt EE, Lamb J, Yang X, et al. An integrative genomics approach to
32. Berezikov E, Guryev V, van de Belt J, Wienholds E, Plasterk R, Cuppen infer causal associations between gene expression and disease. Nat
E. Phylogenetic shadowing and computational identification of human Genet. 2005;37:710 –717.
microRNA genes. Cell. 2005;120:21–24. 56. Zuk O, Hechter E, Sunyaev SR, Lander ES. The mystery of missing
33. Friedman RC, Farh KK, Burge CB, Bartel DP. Most mammalian heritability: genetic interactions create phantom heritability. Proc Natl
mRNAs are conserved targets of microRNAs. Genome Res. 2009;19: Acad Sci U S A. 2012;109:1193–1198.
92–105. 57. Li H, Xuan J, Wang Y, Zhan M. Inferring regulatory networks. Front
34. Shirdel EA, Xie W, Mak TW, Jurisica I. NAViGaTing the micronome: Biosci. 2008;13:263–275.
using multiple microRNA prediction databases to identify signalling 58. Gardner TS, di Bernardo D, Lorenz D, Collins JJ. Inferring genetic
pathway-associated microRNAs. PLoS One. 2011;6:e17429. networks and identifying compound mode of action via expression
35. Loscalzo J, Barabasi AL. Systems biology and the future of medicine. profiling. Science. 2003;301:102–105.
Wiley Interdiscip Rev Syst Biol Med. 2011;3:619 – 627. 59. Lorenz DR, Cantor CR, Collins JJ. A network biology approach to aging
36. Goh KI, Cusick ME, Valle D, Childs B, Vidal M, Barabasi AL. The in yeast. Proc Natl Acad Sci U S A. 2009;106:1145–1150.
human disease network. Proc Natl Acad Sci U S A. 2007;104: 60. Tegner J, Yeung MK, Hasty J, Collins JJ. Reverse engineering gene
8685– 8690. networks: integrating genetic perturbations with dynamical modeling.
37. Dudley JT, Tibshirani R, Deshpande T, Butte AJ. Disease signatures are Proc Natl Acad Sci U S A. 2003;100:5944 –5949.
robust across tissues and experiments. Mol Syst Biol. 2009;5:307. 61. Modi SR, Camacho DM, Kohanski MA, Walker GC, Collins JJ. Func-
38. Reimand J, Tooming L, Peterson H, Adler P, Vilo J. GraphWeb: mining tional characterization of bacterial sRNAs using a network biology
heterogeneous biological networks for gene modules with functional approach. Proc Natl Acad Sci U S A. 2011;108:15522–15527.
significance. Nucleic Acids Res. 2008;36(Web Server issue): 62. Ergun A, Lawrence CA, Kohanski MA, Brennan TA, Collins JJ. A
W452–W459. network biology approach to prostate cancer. Mol Syst Biol. 2007;3:82.
39. Loscalzo J, Kohane I, Barabasi AL. Human disease classification in the 63. Guido NJ, Wang X, Adalsteinsson D, McMillen D, Hasty J, Cantor CR,
postgenomic era: a complex systems approach to human pathobiology. Elston TC, Collins JJ. A bottom-up approach to gene regulation. Nature.
Mol Syst Biol. 2007;3:124. 2006;439:856 – 860.

Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014


374 Circulation Research July 20, 2012

64. Loscalzo J. Personalized cardiovascular medicine and drug devel- 87. Kohanski MA, Dwyer DJ, Wierzbowski J, Cottarel G, Collins JJ. Mis-
opment: time for a new paradigm. Circulation. 2012;125:638 – 645. translation of membrane proteins and two-component system activation
65. Hansen J, Zhao S, Iyengar R. Systems pharmacology of complex trigger antibiotic-mediated cell death. Cell. 2008;135:679 – 690.
diseases. Ann N Y Acad Sci. 2011;1245:E1–E5. 88. Munger J, Bennett BD, Parikh A, Feng XJ, McArdle J, Rabitz HA,
66. Campillos M, Kuhn M, Gavin AC, Jensen LJ, Bork P. Drug target Shenk T, Rabinowitz JD. Systems-level metabolic flux profiling iden-
identification using side-effect similarity. Science. 2008;321:263–266. tifies fatty acid synthesis as a target for antiviral therapy. Nat Biotechnol.
67. Pammolli F, Magazzini L, Riccaboni M. The productivity crisis in 2008;26:1179 –1186.
pharmaceutical R&D. Nat Rev Drug Discov. 2011;10:428 – 438. 89. Warburg O. On the origin of cancer cells. Science. 1956;123:309 –314.
68. Ashley EA, Butte AJ, Wheeler MT, et al. Clinical assessment incorpo- 90. Vander Heiden MG, Christofk HR, Schuman E, Subtelny AO, Sharfi H,
rating a personal genome. Lancet. 2011;375:1525–1535. Harlow EE, Xian J, Cantley LC. Identification of small molecule inhib-
69. Suhre K, Shin SY, Petersen AK, et al. Human metabolic individuality in itors of pyruvate kinase M2. Biochem Pharmacol. 2009;79:1118 –1124.
biomedical and pharmaceutical research. Nature. 2011;477:54 – 60. 91. Michelakis ED, Sutendra G, Dromparis P, Webster L, Haromy A, Niven
70. Sorger PK, Allerheiligen SRB, Abernethy DR, et al. Quantitative and E, Maguire C, Gammer TL, Mackey JR, Fulton D, Abdulkarim B,
systems pharmacology in the post-genomic era: new approaches to McMurtry MS, Petruk KC. Metabolic modulation of glioblastoma with
discovering drugs and understanding therapeutic mechanisms. An NIH
dichloroacetate. Sci Transl Med. 2010;2:31ra34.
White Paper by the QSP Workshop Group; October, 2011. Bethesda,
92. Wise DR, Thompson CB. Glutamine addiction: a new therapeutic target
MD: NIH; 2011:1– 47.
in cancer. Trends Biochem Sci. 2010;35:427– 433.
71. Zanders ED. Overview of chemical genomics and proteomics. Methods
93. Kreeger PK, Lauffenburger DA. Cancer systems biology: a network
Mol Biol. 2011;800:3–10.
modeling perspective. Carcinogenesis. 2009;31:2– 8.
72. Antman E, Weiss S, Loscalzo J. Systems pharmacology, pharmacoge-
netics, and clinical trial design in network medicine. Wiley Interdiscip 94. Laubenbacher R, Hower V, Jarrah A, Torti SV, Shulaev V, Mendes P,
Rev Syst Biol Med. 2012;4:367–383. Torti FM, Akman S. A systems biology view of cancer. Biochim
73. Lamb J, Crawford ED, Peck D, et al. The Connectivity Map: using Biophys Acta. 2009;1796:129 –139.
gene-expression signatures to connect small molecules, genes, and 95. Wellen KE, Lu C, Mancuso A, Lemons JM, Ryczko M, Dennis JW,
disease. Science. 2006;313:1929 –1935. Rabinowitz JD, Coller HA, Thompson CB. The hexosamine biosyn-
74. Hu G, Agarwal P. Human disease-drug network based on genomic thetic pathway couples growth factor-induced glutamine uptake to
expression profiles. PLoS One. 2009;4:e6536. glucose metabolism. Genes Dev. 2010;24:2784 –2799.
75. Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI 96. Gaglio D, Metallo CM, Gameiro PA, Hiller K, Danna LS, Balestrieri C,
gene expression and hybridization array data repository. Nucleic Acids Alberghina L, Stephanopoulos G, Chiaradonna F. Oncogenic K-Ras
Res. 2002;30:207–210. decouples glucose and glutamine metabolism to support cancer cell
76. Chiang AP, Butte AJ. Systematic evaluation of drug-disease rela- growth. Mol Syst Biol. 2011;7:523.
tionships to identify leads for novel drug uses. Clin Pharmacol Ther. 97. Yeh P, Tschumi AI, Kishony R. Functional classification of drugs by
2009;86:507–510. properties of their pairwise interactions. Nat Genet. 2006;38:489 – 494.
77. Suthram S, Dudley JT, Chiang AP, Chen R, Hastie TJ, Butte AJ. 98. Torella JP, Chait R, Kishony R. Optimal drug synergy in antimicrobial
Network-based elucidation of human disease similarities reveals treatments. PLoS Comput Biol. 2010;6:e1000796.
common functional modules enriched for pluripotent drug targets. PLoS 99. Berger SI, Ma’ayan A, Iyengar R. Systems pharmacology of arrhyth-
Comput Biol. 2010;6:e1000662. mias. Sci Signal. 2010;3:ra30.
78. Gottlieb A, Stein GY, Ruppin E, Sharan R. PREDICT: a method for 100. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-driven prediction
inferring novel drug indications with application to personalized of drug effects and interactions. Sci Transl Med. 2012;4:125ra31.
medicine. Mol Syst Biol. 2011;7:496. 101. Xie L, Li J, Bourne PE. Drug discovery using chemical systems biology:
79. Loging W, Harland L, Williams-Jones B. High-throughput electronic identification of the protein-ligand binding network to explain the side
biology: mining information for drug discovery. Nat Rev Drug Discov. effects of CETP inhibitors. PLoS Comput Biol. 2009;5:e1000387.
2007;6:220 –230. 102. Nissen SE, Tardif JC, Nicholls SJ, Revkin JH, Shear CL, Duggan WT,
80. Li J, Zhu X, Chen JY. Building disease-specific drug-protein connec- Ruzyllo W, Bachinsky WB, Lasala GP, Tuzcu EM. Effect of torcetrapib
tivity maps from molecular interaction networks and PubMed abstracts. on the progression of coronary atherosclerosis. N Engl J Med. 2007;
PLoS Comput Biol. 2009;5:e1000450. 356:1304 –1316.
81. Deftereos SN, Andronis C, Friedla EJ, Persidis A. Drug repurposing and
103. Barter PJ, Caulfield M, Eriksson M, et al. Effects of torcetrapib in
adverse event prediction using high-throughput literature analysis. Wiley
patients at high risk for coronary events. N Engl J Med. 2007;357:
Interdiscip Rev Syst Biol Med. 2011;3:323–334.
2109 –2122.
82. Achenbach J, Tiikkainen P, Franke L, Proschak E. Computational tools
104. Lage K, Mollgard K, Greenway S, et al. Dissecting spatio-temporal
for polypharmacology and repurposing. Future Med Chem. 2011;3:
protein networks driving human heart development and related dis-
961–968.
orders. Mol Syst Biol. 2010;6:381.
83. Sirota M, Dudley JT, Kim J, Chiang AP, Morgan AA, Sweet-Cordero A,
Sage J, Butte AJ. Discovery and preclinical validation of drug indi- 105. Sobie EA, Lee YS, Jenkins SL, Iyengar R. Systems biology: biomedical
cations using compendia of public gene expression data. Sci Transl Med. modeling. Sci Signal. 2011;4:tr2.
2011;3:96ra77. 106. Loscalzo J, Kohane I, Barabasi A. Human disease classification in the
84. Dudley JT, Sirota M, Shenoy M, Pai RK, Roedder S, Chiang AP, postgenomic era: a complex systems approach to human pathobiology.
Morgan AA, Sarwal MM, Pasricha PJ, Butte AJ. Computational repo- Mol Syst Biol. 2007;3:124.
sitioning of the anticonvulsant topiramate for inflammatory bowel 107. Chan SY, White K, Loscalzo J. Deciphering the molecular basis of
disease. Sci Transl Med. 2011;3:96ra76. human cardiovascular disease through network biology. Curr Opin
85. Lee MJ, Ye AS, Gardino AK, Heijink AM, Sorger PK, Macbeath G, Cardiol. 2012;27:202–209.
Yaffe MB. Sequential application of anticancer drugs enhances cell 108. Morel NM, Holland JM, van der Greef J, Marple EW, Clish C, Loscalzo
death by rewiring apoptotic signaling networks. Cell. 2012;149: J, Naylor S. Primer on medical genomics, part XIV: introduction to
780 –794. systems biology: a new approach to understanding disease and
86. Kohanski MA, Dwyer DJ, Hayete B, Lawrence CA, Collins JJ. A treatment. Mayo Clin Proc. 2004;79:651– 658.
common mechanism of cellular death induced by bactericidal anti- 109. Schadt EE, Friend SH, Shaywitz DA. A network view of disease and
biotics. Cell. 2007;130:797– 810. compound screening. Nat Rev Drug Discov. 2009;8:286 –295.

Downloaded from http://circres.ahajournals.org/ by guest on May 9, 2014

You might also like