Sie sind auf Seite 1von 16

Cardiovascular Toxicities of Small Molecule

Tyrosine Kinase Inhibitors: An Opportunity for


Systems-Based Approaches
S-A Brown1, L Nhola1 and J Herrmann1

Once universally considered a rapidly fatal condition, cancer has increasingly become a chronic medical condition and
comorbidities and adverse effects of cancer therapies have become increasingly significant. One of the leading
advancements that has gained traction for the treatment of a variety of malignancies is the class of small molecule
tyrosine kinase inhibitors (TKIs). Although, in many aspects revolutionary, TKIs have their own profile of side effects,
including cardiovascular side effects, the most common being hypertension (HTN), congestive heart failure, corrected
QT (QTc) prolongation, and instances of premature coronary heart disease. Herein, we describe the mechanisms of
small TKI-induced cardiovascular toxicity and related intracellular signaling. In particular, systems-based approaches to the
understanding of small TKI-induced cardiovascular toxicity are addressed. The pathophysiology of synergic cardiovascular
toxicity from TKIs, anthracyclines, and monoclonal antibodies (e.g., trastuzumab, bevacizumab) is illustrated. Finally,
recommendations are made for implementing systems medicine in clinical practice, for individualized cardiovascular
wellness after cancer therapy.

Cancer-related mortality rates have been declining since the helped to guide the development of more selective oncologic
1990s, boosted by the development of so-called targeted thera- agents, to predict which patients will be at greatest risk of cardio-
pies.1 The intention was that these drugs would be directed vascular toxicity, and to develop protective strategies to minimize
against a molecular target perceived to be of crucial signicance cardiovascular toxicity.2,4 Herein, we will reect on how systems-
for a malignant disease process. Prime examples are trastuzumab, based approaches, such as genomics, epigenomics, transcriptomics,
a monoclonal antibody to the human epidermal receptor microRNA regulomics, proteomics, metabolomics, microbiomics,
(HER)2, and imatinib, a tyrosine kinase inhibitor (TKI) of and mathematical and computational modelling can be of further
BCR-Abl, the oncogenic fusion product of the Philadelphia chro- value in this regard. These technological tools can provide clues
mosome. These two drugs are, in fact, the front-runners, the rst about variations in different cardiovascular system components in
representatives of the two leading subclasses of targeted therapies response to perturbation by TKI therapy, and how these variations
monoclonal antibodies and small molecule TKIs. Contrary to can affect the pathophysiological phenotype of the cardiovascular
initial expectations, as assumed to be more specic to cancer cells system.
than nontargeted therapies, some of the targeted agents were
found to be associated with signicant cardiovascular morbidity Small molecule tyrosine kinase inhibitor-induced
and mortality.2 Indeed, even in the current era, cancer survivors cardiovascular toxicity
who develop cardiovascular disease have signicantly worse out- Tyrosine kinases (TKs; as part of cellular receptors (receptor
comes compared with cancer survivors without cardiovascular tyrosine kinases (RTKs)) or individual molecules (nonreceptor
disease.3 TKs) regulate a wide array of signaling pathways that control cell
As illustrated for trastuzumab, likely one of the most well- growth, differentiation, migration, apoptosis, and survival. TKs
known targeted therapies associated with cardiotoxicity, elaborate are enzymes that catalyze the transfer of phosphate from adeno-
research efforts revealed that shared biologic pathways account sine triphosphate (ATP) to tyrosine residues of cellular proteins.
for the impact of targeted oncologic therapies on both cancer They thereby not only regulate but even more coordinate the
and the cardiovascular system.4 Such understanding has also function of these proteins as TKs are operating in the broader
1
Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA. Correspondence: J Herrmann (herrmann.joerg@mayo.edu)
Received 30 September 2016; accepted 31 October 2016; advance online publication 2 November 2016. doi:10.1002/cpt.552

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 65


Figure 1 Small tyrosine kinase inhibitor (TKI)-induced cardiotoxicity. Major components of intracellular signaling implicated in TKI-induced cardiotoxicity,
adapted from http://www.nexavar.com/ based on ref. 6 ERK, extracellular signal-regulated kinase; FGFR, fibroblast growth factor receptor; MAPK,
mitogen-activated protein kinase; MEK, methyl ethyl ketone; PDGF, platelet-derived growth factor; PDGFR, platelet-derived growth factor receptor; RAS,
renin angiotensin system; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.

framework of signaling pathways governed by extracellular stimu- phosphoinositide 3 kinase-AKT and RAS-RAF-mitogen-
li or ligands, such as growth factors (e.g., vascular endothelial activated protein kinase (MAPK)/extracellular signal-regulated
growth factor (VEGF) and platelet-derived growth factor kinase-extracellular signal-regulated kinase pathways, as illustrated
(PDGF)) or intracellular stimuli, such as oxidant stress (Figure 1).5,6 in Figure 1.6,9
Hyperactivation of TK signaling has been shown to play a role in Related to these mechanisms, hypertension (HTN) has
the development of cancer and tumor angiogenesis, thus generating emerged as the most common cardiovascular side effect of TKI.9
an attractive venue for drug development.5 In addition to HTN, left ventricular dysfunction and heart fail-
Importantly, most TKIs compete with ATP for binding to the ure (HF) have been associated with small-molecule multi-TKI
ATP-binding domain (or ATP-binding pocket), which is a rela- therapy.2,10 The incidence of left ventricular dysfunction and HF
tively conserved structure across several kinases, and for this rea- varies in the literature (233%), which, in part, is due to differ-
son lack 100% specicity for their intended molecular target.4,5 ences in patient populations, drug regimens, and a lack of consen-
This leads to inhibition of intended tumor pathways, which is sus over denitions for cardiovascular toxicity (estimates for
benecial in cancer therapy, but also inhibits off-target kinases.5 different TKIs are listed in Table 1).2,10
Although these off-target kinases may or may not have a role in The specic molecular mechanisms underlying the develop-
cancer cell survival and proliferation, they may be crucial to the ment of cardiomyopathy with small molecule TKIs have not
survival and function of other cells (e.g., cardiomyocytes), with been fully elucidated and varies based on the inhibitory spectrum
important clinical implications (i.e., off-target effects).5 of the individuals TKIs. Sunitinib, for instance, inhibits at least
A number of multitargeted TKIs have been approved by the 50 kinases, generating a high level of complexity. Prior studies,
US Food and Drug Administration, such as sunitinib, sorafenib, however, outlined mitochondrial damage, energy rundown, and
pazopanib, axitinib, vandetanib, cabozantinib, ponatinib, and off target impairment of the energy-restoring activity of AMP-
regorafenib, whose common target is interference with VEGF sig- activated protein kinase (AMPK).11 Importantly, AMPK was
naling (Table 1).7 Although this is of signicance for tumor inhibited with an IC50 of 216 6 58 nM, comparable to other
angiogenesis and growth, VEGF signaling is of crucial importance molecular targets of sunitinib and at a concentration level that
for normal cardiovascular function.8 This pertains primarily to matches trough blood levels of sunitinib plus its major active
endothelial cells, in terms of proliferation, survival, and function. metabolite, which are in the order of 125250 nM.12
Induction of endothelial cell death and rarefaction of resistance Recently, the focus shifted to the primary targets of sunitinib,
vessels can be a consequence, as well as impaired vasodilation.9 namely VEGF and PDGF. Mice with cardiac or endothelial cell-
Regulation of nitric oxide (NO) production is an important ele- specic deletion of VEGF develop capillary rarecation, induc-
ment, intertwined with VEGF receptor signaling via the tion of hypoxia-inducible genes, and cardiomyopathy. Similarly,

66 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


Table 1 Small molecule tyrosine kinase inhibitors (modified from ref. 7)
Stroke/
Malignancy targets Molecular targets HTN Chest pain MI TIA PAD DVT/PE Additional warnings
Bcr/Abl inhibitors
Imatinib Ph1 CML, Ph1 ALL, ABL-1, c-KIT, PDGFR-A, 4% 711% Edema 1186%
GIST, ASM, MDS PDGFR-B, FLT-3 Hypotension 11%
Off-label melanoma
Nilotinib Ph1 CML ABL-1, c-KIT, PDGFR-A, 1011% 59% 13% 34% Pericardial effusion 2%
Off-label GIST PDGFR-B, FLT-3
Ponatinib Ph1 CML, Ph1 ALL ABL-1, c-KIT, PDGFR-A, 5371% 12% 2% 8% 5% HF 615%
PDGFR-B, FLT-1, KDR, Pericardial effusion 13%
FLT-3, FLT-4, FGFR, Atrial fibrillation 4%
SRC, TIE2
Bosutinib Ph1 CML ABL-1, FGFR, FLT-1, Edema 14%
FLT-2, FLT-3, KDR, Pericardial effusion 13%
PDGFR-A, PDGFR-B, SRC
Dasatinib Ph1 CML, Ph1 ALL ABL-1, FGFR2, KIT, 4% IHD Edema 4%
Off-label GIST PDGFR-A, PDGFR-B, SRC Pericardial effusion 4%
Cardiac dysfunction 4%
QTc prolongation 1%
Conduction
abnormalities 7%

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017


VEGFR inhibitors
Sorafenib RCC, thyroid cancer, B-Raf, FLT-1, FLT-3, FLT-4, 941% 3% 1% HF in 2%
HCC, off-label: KDR, KIT, PDGFR-A, (grades 3
angiosarcoma PDGFR-B, FGFR, c-fms or 4 10%)
and GIST
Sunitinib RCC, GIST, PNET, ABL-1, c-KIT, PDGFR-A, 2734% 13% 3% each LVEF drop 1627%
off-label: STS, PDGFR-B, FLT-1, KDR, (grade 3
thyroid cancer FLT-3, FLT-4, FGFR, or 4 10%)
SRC, c-smc
Pazopanib RCC, STS, thyroid ABL-1, c-KIT, PDGFR-A, 40% (grade 510% 2% 1% 15% HF 11%
cancer PDGFR-B, FLT-1, KDR, 3 or 4 7%) QTc prolongation 2%
FLT-4, FGFR, c-fms Bradycardia 219%
Axitinib RCC c-KIT, PDGFR-A, PDGFR-B, 40% (grade 2% 1% 13%
FLT-1, KDR, FLT-4, 3 or 4 16%)
Regorafenib GIST, colorectal PDGFR-B, FLT-1, KDR, 3059% 1%
cancer FLT-4, TIE2, RET, (grade 3 or
c-KIT, RAF 4 828%)
Lenvatinib Thyroid cancer PDGFR-B, FLT-1, KDR, 73% (grade 3% LVEF drop 2%
FLT-4, RET, c-KIT 3 or 4 44%) QTc prolongation 9%
Table 1 Continued on next page

67
68
Table 1 Continued

Stroke/
Malignancy targets Molecular targets HTN Chest pain MI TIA PAD DVT/PE Additional warnings
EGFR inhibitors
Erlotinib NSCLC, pancreatic EGFR (HER1) N/a 12% (2%) (3%) N/a (4%) (Cardiac arrhythmia 5%,
(1gemcitabine) cancer syncope 5%,
thrombosis 11%)
Vandetanib Thyroid cancer EGFR, KDR, 33% (grade 1% QTc prolongation 14%
FLT-4, RET 3 or 49%) Heart failure 2%
ALK inhibitors
Crizotinib NSCLC ALK, MET, 6% QTc prolongation 56%
ROS1 Bradycardia 515%
Syncope 13%
Edema 3149%
Ceritinib NSCLC ALK, IGF-1R, QTc prolongation 4%
ROS1, InsR Bradycardia 3%
JAK inhibitors
Ruxolitinib Myelofibrosis JAK1, JAK2, 6%
Polycythemia TYK2
vera
ALK, anaplastic lymphoma kinase; ALL, acute lymphoblastic leukemia; ASM, aggressive systemic mastocytosis; CML, chronic myeloid leukemia; DVT, deep vein thrombosis; FGFR, fibroblast growth factor receptor; FLT, Fms-
like tyrosine kinase; GIST, gastrointestinal stromal tumor; HCC, hepatocellular carcinoma; HER, human epidermal receptor; HF, heart failure; HTN, hypertension; IHD, ischemic heart disease; LVEF, left venrticular ejection
fraction; MDS, myelodysplastic syndrome; MET, mesenchymal-epithelial transition factor; MI, myocardial infarction; N/A, not applicable; NSCLC, nonsmall cell lung cancer; PAD, peripheral arterial disease; PDGFR, platelet-
derived growth factor receptor; PE, pulmonary embolism; Ph1, Philadelphia chromosome-positive; PNET, pancreatic neuroendocrine tumors; QTc, corrected QT.

VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


deletion of the PDGF receptor beta leads to decreased capillary sunitinib-induced cardiovascular toxicity (e.g., HTN, arterial
density, hypoxia, and cardiac dysfunction and remodeling upon thromboembolic events) have been discovered1528 (Figure 2, top
pressure overload. This has led to the theory that induction of hyp- left), and are described in the following sections. Interestingly, the
oxia and hypoxia response elements is the underlying mechanism variants are relevant to almost every major step of sunitinib phar-
of cardiomyopathy observed with TKIs that target the VEGF and macodynamics or pharmacokinetics. Many variants are related to
PDGF signaling pathway. Accordingly, the presentation would be transcription factors that bind VEGF promoter regions.15
more consistent with myocardial hibernation.4 Indeed, ultrastruc-
tural changes, including those of the mitochondria, and cardiac dys- Pharmacokinetics
function are reversible in most cases of sunitinib cardiotoxicity. Absorption/efflux. The ATP-binding cassette transporter protein
Moreover, a recent study pointed out the detrimental role of peri- ABCG2 mediates absorption and/or excretion of sunitinib
cyte depletion via inhibition of PDGF-beta signaling upon micro- through the wall of the intestine and also efux of sunitinib from
vascular and cardiac function.13 This has generated the concept of varied cells targeted by the drug.17 The ABCG2 variant 421C>A
pericyte-endothelial-myocardial coupling as an important element (rs2231142) associates with high intracellular concentrations of
in the induction of small molecule TKI-induced cardiotoxicity, as sunitinib, with consequent adverse events, such as HTN.18 The
illustrated in the example of sunitinib.14 ABCG2 TT haplotype (15622C/T and 1143C/T; rs2622604)
associates with grade >2 sunitinib-induced toxicities, including
Systems-based approaches HTN.19 The ABCB1 variants rs1128503C>T (variation
Cardiovascular toxicity is mainly a pleiotropic or off-target effect G412G) and rs2032582G>T (variation A893S) show a trend
of small molecule TKIs, for which every patient treated with toward protection from sunitinib-induced HTN.16
small molecule TKIs should be at risk. Yet, there is variability in
observed cardiovascular toxicity. This suggests that there may be Metabolism. The CYP3A5 variant rs776746 associates with dose
unique characteristics of genetics and interaction with lifestyle reductions due to grade 2 toxicities, including HTN.20 This
and therapeutic environment that result in a small percentage of variant at the CYP3A5*1 allele is thought to increase CYP3A5
individuals experiencing HTN, congestive heart failure, prema- expression, thereby increasing conversion of sunitinib to its more
ture coronary heart disease, prolonged corrected QT (QTc), or metabolite SU12662.21 The metabolite has a longer half-life than
other forms of cardiovascular toxicity. Systems-based approaches sunitinib, and is thought to therefore lead to increased exposure
to evaluating cardiovascular toxicity in response to small molecule to the effect of sunitinib therapy.21,22
TKI therapy could accelerate understanding of pathophysiology
and development of safe and effective drugs. Systems-based Pharmacodynamics
approaches are hallmarks of systems biology, which uses biotech- Receptor substrate. The vascular endothelial growth factor recep-
nological tools to study whole cells, tissues, or organisms. Systems tor type 2 (VEGFR2) receptor variant 1191T>C (rs2305948)
biology studies integrate disparate cellular processes and signaling also associates with grade >2 sunitinib-induced toxicities, including
pathways at different levels of the organism, and in multiple HTN.19 The VEGFR2 variant rs1870377T>A (variation Q472H)
dimensions. Systems medicine is the application of systems biolo- trends toward association with sunitinib-induced HTN.16
gy to elds of medicine, including cardio-oncology.
The mechanisms of cardiovascular toxicity from small mole- Receptor ligand substrate. The VEGF variant -634G>C
cule TKI therapy are not yet fully understood. Reviewing the (rs2010963) associates with sunitinib-induced HTN.23 The
current state of our ndings from systems medicine tools may help VEGF variant rs1570360 associates with increased risk for
set the stage for further analyses to inspire inferences about the sunitinib-induced HTN.20 The VEGF-A variant rs699947A>C
steps underpinning toxicity. In the following sections, we review (promoter) trended toward association with sunitinib-induced
systems-based approaches to studying genomic, transcriptomic, HTN.16 In addition, the VEGF variants -1498C>T (rs833061)
pathway, and other variations resulting from perturbation of the and -634G>C (rs2010963) associates with changes in VEGF
cardiovascular system by TKI therapy. Integrating such data from expression levels and bevacizumab-induced HTN.29,30 In fact,
diverse systems medicine tools could potentially advance our under- the variant -634G>C (rs2010963) is critical for expression of a
standing of the pathophysiology of TKI-induced cardiotoxicity and VEGF isoform.31
point to high-yield effective therapeutic avenues. Ultimately, incor- The interleukin-8 variant rs1126647A>T associates with
porating these systems-level data into individualized comprehensive sunitinib-induced HTN,24 presumably by increasing interleukin-
predictive modeling should help personalize the study of these per- 8 expression levels,25 which may upregulate VEGF levels through
turbations for each patient as a whole system. As HTN is the most NFkappaB.26,27 The interleukin-13 variant rs1800925C>T also
common cardiovascular adverse effect of inhibitors of the VEGF associates with grade >2 sunitinib-induced toxicities, including
pathway,15,16 most studies in the following sections focus on HTN HTN.24
as the main form of toxicity.
Downstream effector. In response to stimulation of VEGFR2,
Genomics NO is produced.32,33 This is mediated by promoting expression
For simplicity, this section focuses on cardiovascular toxicity of NO synthases,34 such as endothelial NO synthase, which is
from sunitinib therapy. Several genomic variants underlying found in endothelial cells and cardiomyocytes, among other cell

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 69


Figure 2 A systems medicine approach to prediction, prevention, and therapy. Left: Numerous systems biology tools are available for use in cardio-
oncology, to yield and analyze systems medicine data, such as results from genomic, transcriptomic, metabolomics, network medicine, and computational
biology assessments. Right: Patients with cancer will have the capability to undergo testing with a number of systems medicine technologies. Test results
will suggest the patients individual probabilistic low or high risk for developing cardiotoxicity. A therapeutic plan should then be devised to minimize other
contributors to risk, such as concomitant hormonal or radiotherapy. Protective cardiovascular drugs could also be initiated, guided by the patients likeli-
hood of being a responder or nonresponder to this medication. Sunitinib cardiotoxicity genetic variants not shown are: ABCB1, ATP binding cassette sub-
family B member 1 (efflux pump/transporter); ABCG2, ATP-binding cassette sub-family G member 2 (efflux pump/transporter); CYP3A5, cytochrome P450
family 3 subfamily A member 5 (pharmacokinetics/metabolism); IL8/13, interleukin-8/interleukin-13 (inflammation/cytokine); NR1I2, nuclear receptor
subfamily 1, group I, member 2 (nuclear transcription/localization, regulates CYP3A and ABCB1); VEGFA, vascular endothelial growth factor A
(angiogenesis).

types.35 In concert with prostaglandins, endothelial cell-dependent an afnity for areas of the RTK that are located outside of the
vasodilation of the arterial and venous systems ensue to control ATP-binding site.36 These TKIs modify the RTKs tridimension-
blood pressure.15,32,33 Consistent with this, the variant endothelial al structures and disrupt the ATP-kinase pocket interaction. Oth-
nitric oxide synthase rs2070744 associates with grade 3 sunitinib- er TKIs function as covalent inhibitors. These have an afnity for
induced HTN.28 the ATP-binding site of RTKs, but bind irreversibly by covalently
bonding.36 As a result, a generalized class effect cannot be inferred
for the inuence of small molecule TKIs on outcome, efcacy, or
Inherent differences among agents
There are inherent differences between agents,15 as they have dif- cardiovascular toxicity.37
ferent interaction sites on their substrates, which affects potency Variant overlap for toxicity and outcome/efficacy
and afnity of binding (e.g., to VEGFR).9,15 Therefore, a variant Authors have noted overlap in variants that associate with toxici-
associated with toxicity from one drug may not associate toxicity ty and outcome or efcacy. As one might expect, variants that
from another. For example, the VEGF variants -1498C>T associate with more toxicity often also associate with more cyto-
(rs833061) associates with bevacizumab-induced HTN, but not toxic efcacy or better outcome in terms of progression-free sur-
sunitinib-induced HTN.15 Additionally, TKIs can be categorized vival or overall survival, and the converse is also true.15,16
into at least four main groups.36 Some TKIs are ATP-competitive
inhibitors. These have an afnity for the ATP-binding site of Transcriptomics
RTKs in their active conformation.36 Some TKIs have an afnity Gene expression proles, or transcriptomes, have been used to
for the ATP-binding site of RTKs in their nonactive conforma- uncover pathways involved in cardiovascular toxicity. For exam-
tion.36 In this case, activation of the RTK becomes energetically ple, results from a transcriptomics study illustrated the role of
unfavorable. Some TKIs function as allosteric inhibitors and have AMPK in survival of cardiomyocytes during small molecule TKI

70 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


therapy.38 Analysis of gene expression patterns indicated that a can be indicative of metabolism, stress, and injury, and cardiac
lapatinib analog led to activation of AMPK, with subsequent magnetic resonance imaging and 18-uorodeoxyglucose positron
effects on p53 and apoptosis. Although lapatinib has overall car- emission tomography can detect energetic alterations, but the
diotoxic effects, this pathway seemed to be cardioprotective and latter has not yet been well developed for cost-effective care of
may function as a homeostatic mechanism. This is in contrast to individuals with cardiovascular toxicity,42 or for those at risk of
studies showing that sunitinib-induced cardiovascular toxicity is toxicity.
mediated by direct inhibition of AMPK.12 It is thus important to When assessing the microbiome, a bacteriomic signature was
recognize that different mechanisms underlie toxicity from differ- discovered for individuals undergoing TKI therapy, relative to
ent TKIs, limiting generalizations about class effects. Another healthy individuals.47 DNA analysis of fecal microbiota further
study reviewed assorted pathways (determined in part from tran- revealed a unique bacterial signature for individuals at high vs.
scriptomic studies) that mediate the cardioprotective effects of low risk of developing diarrhea in response to TKI therapy.47
aerobic exercise therapy in the setting of small molecule TKI Potentially, microbiomics could also be used to evaluate and
therapy.39 Some groups have used gene expression patterns to predict TKI-induced cardiovascular toxicity.
establish the utility of distinct cell lines for examining the efcacy Other systems biology tools, such as epigenomics (or methylo-
of small molecule TKI therapy, and suggested that these could mics),48 proteomics,49 interactomics,49,50 network medicine,49 and
also potentially be used to assess mechanisms of cardiovascular mathematical or computational biology49 have also provided useful
toxicity.40 Other groups have used transcriptomics to delineate information regarding mechanisms of adverse drug effects, develop-
signaling pathways in response to RTKs,41 and have established ment of drug targets,50 optimization of drug delivery,51 determi-
cell lines that could potentially also be used to evaluate cardiovas- nants of drug resistance and sensitivity,52 and pathophysiology of
cular toxicity from TKI therapy.42 One such cell line is composed cancers4852 and cardiovascular disease.46,48 Network biology or
of human-induced pluripotent stem cell-derived cardiomyocytes medicine is the consolidation of multiple networks of proteins,
derived from broblasts.41 These cell lines hold great promise for cells, or other units for comprehensive analysis of the relationships
personalizing therapy.43 For instance, a recent study showed that among these entities. Mathematical or computational biology or
pluripotent cells from individuals with breast cancer exhibit dif- medicine is the use of mathematical or computational methods for
ferential gene expression proles if they develop doxorubicin- studying biology or systems in medicine. Several of these tools have
induced cardiovascular toxicity.43 Similar studies could be pur- been combined to enhance investigations.49 These systems medi-
sued to identify a gene expression prole unique to individuals cine tools in diverse combinations are now primed for investigating
who develop TKI-induced cardiovascular toxicity. This could small molecule TKI-induced cardiovascular toxicity. Using systems
then be used to guide therapy in precision medicine. medicine tools, we may approach an era in which optimal dosing
can be selected to minimize cardiovascular toxicity and maximize
Other systems medicine tools efcacy and outcomes.
In addition to genomics and transcriptomics, there are different
systems biology tools that have the potential to someday contrib- SYNERGY OF CHEMOTHERAPEUTICS
ute to the conversation about TKI-induced cardiotoxicity. Three Anthracyclines and monoclonal antibodies to receptor
of these have been associated with ndings in cardiovascular dis- tyrosine kinases
ease or cancer, namely microRNA regulomics, metabolomics, and The prime example of synergy in chemotherapeutic cardiotoxicity
microbiomics. is that of anthracyclines and trastuzumab. First in its class, trastu-
MicroRNAs (or miRNAs) are small (1825 nucleotides), zumab became the frontrunner for mAb-based cancer therapy,
single-stranded, noncoding RNAs that repress translation of especially for patients with cancers that overexpress the HER2.53
mRNAs by binding to imperfectly complementary-specic In keeping with the initially expressed thought that molecularly
mRNA sequences.44,45 The miRNAs that have been found to be targeted therapy would equate highly cancer-specic therapy;
critical for heart function are involved in vascular remodeling, reports of cardiotoxicity came as a complete surprise. This includ-
coronary artery disease, and heart failure.46 Although several stud- ed a 2% incidence of cardiotoxicity with trastuzumab monother-
ies have implicated miRNAs in cancer pathophysiology, no stud- apy, and a 27% incidence of cardiotoxicity when trastuzumab
ies have yet elucidated the role of miRNA regulomics in was combined with anthracyclines.4,54 Trastuzumab is very effec-
cardiovascular toxicity following small molecule TKI therapy. tive in blocking the activation of extracellular signal-regulated
Nevertheless, expression of miRNA-34 has been shown to pro- kinase, phosphoinositide 3 kinase/AKT, and JNK-STAT signal-
mote apoptosis of pro-angiogenic cells and reduce the expression ing pathways in HER2-positive breast cancer, and it was subse-
of Bcl-2 and other anti-apoptotic proteins in patients with car- quently realized that it also interfered with important signaling
diovascular disease.44 The miRNA expression and function may, pathways in the heart. A clinical observation, therefore, led to
therefore, become an attractive area of study in cardiovascular further investigations into the previously unknown nuances of
toxicity from cancer therapy in the near future. HER2 signaling in cardiomyocytes. This signaling pathway is rap-
Metabolic signatures in the blood and strategies to examine idly downregulated in the heart after birth, but is re-expressed
alterations in metabolic states could potentially become useful for under conditions of stress.55 This can include afterload stress,
early identication of cardiovascular toxicity,42 and even for pre- ischemic stress, or chemical stress (e.g., by anthracyclines). Con-
dicting and preventing toxicity. Metabolic proles from blood ceivably, anthracyclines, by their acute accumulation in

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 71


mitochondria, trigger a stress response in cardiomyocytes that from the cell. Inhibition of all three pathways decreases the levels
involves the HER2 signaling pathway and abates over time. or activity of transcription factors that regulate growth, survival,
Indeed, this model explains very well the detrimental effects of and cell cycle progression.59 In addition, inhibition of the MAPK
concomitant exposure and the substantially lower risk of cardio- pathway increases levels of reactive oxygen species (ROS), leading
toxicity with trastuzumab with increasing time from anthracy- to apoptosis. Monoclonal antibodies to RTKs, such as bevacizu-
cline exposure. Moreover, stimulation of this pathway with mab, can synergize disruption of these intracellular signaling
recombinant neuregulin-1, the endogenous ligand of HER2 nor- pathways. Further, upon entering the cell, doxorubicin binds
mally released by cardiac endothelial cells, can rescue cardiomyo- topoisomerase alpha or beta.59 The doxorubicin/topoisomerase
cytes in culture from the myobrillar disarray caused by complex induces double-stranded DNA breaks, inuences tran-
anthracyclines.55 scription in the nucleus, and increases ROS in the mitochondri-
There has also been recent recognition that increases in sympa- on, contributing to mitochondrial dysfunction and apoptosis.59
thetic tone may be a mechanism of HER2-targeted therapy alter- The extent of HTN occurring in response to therapy with
ing cardiac function. In preclinical models, the b-adrenergic VEGF signaling pathway inhibitors is dose-dependent,15 consis-
receptor expression and activity is coupled to HER2 expression56 tent with synergistic activation of downstream effectors leading
and HER2 activation is a critical modulator of toxicity to chronic to amplied toxicity. Thus, small molecule TKIs, anthracyclines,
b-adrenergic receptor agonist exposure.56 Concordantly, patients and monoclonal antibodies to RTKs can stimulate or inhibit
with breast cancer who are treated with HER2-targeted therapy overlapping intracellular signaling pathways with potential for
demonstrated increased levels of norepinephrine along with synergy. This would support ndings suggestive of synergy in
increased blood pressure and heart rate.56 Accordingly, there may clinical trials.
be provocation of the very mechanisms that activates an HER2-
mediated stress response in patients who undergo treatment with MULTIPLE HIT HYPOTHESIS
HER2 inhibitors and increased sensitivity to any concomitant The multiple hit hypothesis was originally developed as a concep-
alternate chemotherapy, especially anthracycline combination tual model to explain carcinogenesis as the consequence of multi-
therapy. ple DNA mutations that, over time, critically alter the expression
and activity of proto-oncogenes and tumor suppressor genes. The
Small molecule tyrosine kinase inhibitors, anthracyclines, and idea was rejuvenated by Jones et al.60 in efforts to provide a
monoclonal antibodies to receptor tyrosine kinases framework for the emerging eld of cardio-oncology. According-
In a clinical trial, patients with histologically proven advanced ly, every patient with cancer presents with a given cardiovascular
hepatocellular carcinoma who were treated with doxorubicin and reserve, which can be reduced by any pre-existing cardiovascular
sorafenib combination therapy vs. doxorubicin monotherapy condition and is challenged by the cancer and its treatments. The
showed improvement in time to progression, overall survival, and very fact that cardiovascular risk factors and diseases are predic-
progression-free survival.57 The trial authors speculated that tive of cardiovascular toxicity is supportive of this idea, although
improvement may have been due to synergism between doxorubi- it has never been directly proven experimentally.
cin and sorafenib.57 Eight patients treated with doxorubicin and An indirect illustration, however, is provided by studies on a
sorafenib combination therapy but not doxorubicin monother- particular class of chemotherapeutics: proteasome inhibitors.61
apy developed HTN. In another study, patients with histological- Although coronary atherosclerosis and cardiac dysfunction was
ly proven advanced hepatocellular carcinoma who were treated also noted in normal animals undergoing proteasome inhibitor
with doxorubicin and sorafenib combination therapy vs. sorafe- treatment, the degree of cardiovascular injury was more profound
nib monotherapy showed no improvement in overall survival, in animals with concomitant hypercholesterolemia. The potential
and progression-free survival.58 Although cardiovascular toxicity explanation was that proteasome activity is upregulated under
was not explicitly delineated, the authors noted grade 3 or 4 stress conditions as a compensatory response for the increase in
hematologic adverse events in 37.8% of individuals receiving proteolytic demand and the consequences of proteasome inhibi-
doxorubicin and sorafenib combination therapy, compared with tion are thus much more severe under these circumstances. Simi-
8.1% in individuals receiving sorafenib monotherapy.58 This sug- larly, one may postulate that a reduction of baseline proteasome
gested synergistic hematologic toxicity in those receiving doxoru- function as seen with aging may pose a greater risk. Thus, where-
bicin and sorafenib combination therapy. It will be interesting to as proteasome inhibition could induce cardiovascular disease as
see a more detailed report of this study to determine whether one single hit depending on dose and duration even under nor-
indeed cardiovascular adverse effects were adequately assessed and mal circumstances, any additional pre-existing or concomitant hit
compared between the two groups. on the proteasome system is an aggravating factor for potential
TKIs inhibit multiple receptors, including VEGFR, platelet- cardiovascular toxicity.
derived growth factor receptor (PDGFR), and the ERBB2/ TKIs are a typical example of cancer therapeutics that by them-
ERBB4 heterodimer (Figure 3). This antagonizes the MAPK, selves can induce multiple hits. Sunitinib, for instance, inhibits
phosphoinositide 3 kinase-AKT, and JAK-STAT intracellular not only VEGF signaling but also PDGF signaling. These multi-
signaling pathways. Inhibition of the MAPK and phosphoinosi- ple hits have multiple profound consequences on the vasculature.
tide 3 kinase-AKT pathways decreases MDR1 availability on the First of all, PDGF-beta, produced by endothelial cells, stimulates
cell membrane.59 This limits efux of drugs, such as doxorubicin an essential survival signaling pathway in pericytes, and VEGF,

72 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


Figure 3 Overlapping intracellular signaling pathways in synergistic cardiotoxicity. Mechanisms of cardiotoxicity resulting from interactions of small
tyrosine kinase inhibitor (TKI) signaling pathways with those of other therapeutics, such as anthracyclines and human epidermal receptor 2 (HER2)
receptor antibodies. HER2 receptor antibodies bind to the HER2 (also known as ERBB2, named for a similarity to the avian erythroblastosis viral
oncogene) and HER4 (also known as ERBB4) heterodimer (ERBB2, ERBB4). HER2/ERBB2 5 human epidermal growth factor receptor 2, HER4/ERBB4 5
human epidermal growth factor receptor 4. MAPK, mitogen-activated protein kinase; PDGFR, platelet-derived growth factor receptor; VEGF, vascular
endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.

produced by pericytes, is an essential signaling pathway for with cancer who are undergoing limited rounds of chemotherapy.
endothelial cell survival and function.14,62 Accordingly, a dual, In distinction, patients receiving radiotherapy sustain concurrent
anti-PDGF and anti-VEGF, effect is much more detrimental hits, similar to the original theory of cancer related to radiation.66
than either one alone, and profound consequences in terms of Cardiovascular risk factors and pre-existing coronary artery dis-
microvascular dysfunction and reduced contractile reserve have ease are potent determinants of ischemic events after chest radio-
been shown.62 Furthermore, there is not only coupling of therapy in individuals with breast cancer.67 These observations as
pericytes and endothelial cells but also of endothelial cells with well as the general considerations outlined herein provide impe-
vascular smooth muscle cells or cardiomyocytes.14 The multiple tus for adoption of a mnemonic: awareness and aspirin, blood
hit theory on the endothelial cell level is further illustrated below. pressure, cholesterol and cigarette use, diet and diabetes mellitus,
As outlined above, endothelial cells produce neuregulin-1, and exercise (ABCDE).68 The mnemonic reects the multifactorial
with their impairment, major alterations of the neuregulin-1 sig- nature of (and hits in) cardiovascular disease and is geared to lim-
naling pathway can be expected as well. Reduction of NO pro- it the extent of injury to minimize any untoward long-term
duction is equally profound, affecting muscular tone, stiffness, consequences.
and pressure.63,64 Accordingly, multitargeted TKIs, like sunitinib,
can have a profound impact on the cardiovascular system. Multiple hits in vascular endothelial cells
With regard to cancer therapies, one can distinguish between In the endothelial cell, TKIs inhibit diverse RTKs, which disrupt
those drugs that induce a fairly limited scope of injury and may regulation of angiogenesis and metabolism, and adaptation to
require additional factors to generate clinical consequences and stress, as well as growth, survival, and homeostasis (Figure 4).
those agents that induce multiple injuries sufcient to become Inhibition of VEGFR also decreases NO levels, which limits
clinically evident. For the vasculature, the response to injury theo- relaxation of adjacent vascular smooth muscle cells (VSMCs) and
ry proposes that atherosclerosis develops as a response to any type contributes to HTN.63,69 As shown in Figure 4, disruption of
of endothelial injury.65 Indeed, endothelial dysfunction is noted regulation of metabolism and adaptation to stress are linked to
frequently under diverse clinical conditions and is the rst step. mitochondrial dysfunction.63,64 The cardiovascular system is
However, it is still reversible, and no permanent damage is critically dependent on normal mitochondrial morphology, distri-
induced until structural changes occur. These are more likely to bution, DNA regulation, and function.63 Mitochondrial dysfunc-
evolve the more severe and the more chronic the exposure. This tion increases levels of cytotoxic ROS. The ROS levels also
may explain why atherosclerosis is not more common in patients increase in the setting of oxidized low density lipoprotein

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 73


Figure 4 Proposed mechanisms for the multiple hit hypothesis. Underlying susceptibility to cardiovascular injury, based on an individuals systems
medicine makeup and any relevant personal/family medical history, may predispose to cardiotoxicity, including premature coronary heart disease, subse-
quent to small molecule tyrosine kinase inhibitor (TKI) therapy. ABCDE, awareness and aspirin, blood pressure, cholesterol and cigarette use, diet and
diabetes mellitus, exercise; PDGF, platelet-derived growth factor; PDGFR, platelet-derived growth factor receptor; RTK, receptor tyrosine kinase; VEGF,
vascular endothelial growth factor; VSMC, vascular smooth muscle cell.

cholesterol, triglycerides, hyperglycemia, and free fatty acids.63 atherosclerosis,63,64 TKI-induced atherosclerosis is likely mediat-
ROS disrupts endothelial nitric oxide synthase, with subsequent ed in part by mitochondrial dysfunction and dysfunctional angio-
reduction in NO levels.63 NO is considered an antihypertensive, genesis. Mitochondrial dysfunction leads to increased production
antithrombotic, and anti-atherosclerotic protein.63 Under normal of ROS and decreased levels of calcium and NO and thereby
conditions, mitochondria also regulates intracellular calcium lev- endothelial dysfunction.63,64 Reduced calcium concentration
els by buffering and releasing calcium near calcium inux or worsens mitochondrial dysfunction, and also independently
release channels, respectively.63 This mitochondrial regulation of decreases NO levels further.63,64 Decreased levels of NO impair
calcium modulates cell metabolism and survival, and also mito- vascular relaxation and aggravate HTN, perpetuating the dynam-
chondrial biogenesis.63 Besides TKI therapy and other causes, ics.70 ROS worsens mitochondrial dysfunction, and also leads to
doxorubicin therapy also contributes to mitochondrial dysfunc- VSMC dysfunction, adhesion, and activation of inammatory
tion.59 Mitochondrial dysfunction then results in dysfunction of cells, and macrophage apoptosis, all of which potently pro-athero-
VSMCs and adhesion and activation of inammatory cells.64 genic in effects.63,64,71
The outcome of these concurrent and collaborative processes is Close inspection of the TKI signaling pathways that lead to
atherosclerosis. Patient characteristics, such as age, personal medi- atherosclerosis reveal a myriad of potential feedback and feedfor-
cal history, and family history, also contribute to atherosclerosis. ward loops. Positive feedforward loops propagate the signal to a
Modiable cardiovascular risk factors captured by the mnemonic particular downstream protein through two separate and comple-
ABCDE can also contribute, as host susceptibility and character- mentary signaling pathways, whereas negative feedforward loops
istics help to determine development of atherosclerosis. Other activate a downstream protein in one arm and block activity in
cardiotoxic therapies (e.g., radiation), can also impact preserva- the same downstream protein in the other arm of the loop. Simi-
tion of growth, survival, or homeostasis. larly, positive feedback loops strengthen the signals from
upstream proteins, whereas negative positive feedback loops
Feedback/feedforward loops antagonize the signals from upstream proteins.
Figure 5 shows the substrate for several potential feedback and TKI-induced atherosclerosis potentially boasts at least 10 posi-
feedforward loops involved in cardiovascular toxicity resulting tive feedforward loops and 2 positive feedback loops. Eight of
from small molecule TKI therapy, particularly premature coro- these feedforward loops involve HTN. All of these feedback and
nary heart disease. As in other circumstances of feedforward loops are identied in Figure 5. Several loops, which

74 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


Figure 5 Potential feedback and feedforward loops inherent in small molecule tyrosine kinase inhibitor (TKI) cardio-oncology. Small molecule TKI therapy
may function as a molecular switch to turn on premature atherosclerosis by inducing several positive feedforward loops (1FFLs). Each 1FFL has two
arms. One arm proceeds from X to Z, with X leading to an impact on Z (upper left inset); the second arm proceeds from X to Y to Z, with X impacting Y,
which also impacts Z. In this way, X influences Z through two separate and complementary pathways. In small molecule TKI cardio-oncology,
mitochondrial dysfunction is a conduit for many of these 1FFLs, along with positive feedback loops (1FBLs). These 1FFLs and 1FBLs subsequent to
mitochondrial dysfunction may also be common to anthracyclines and monoclonal antibodies to receptor tyrosine kinases (RTKs). However, the
anti-angiogenesis pathway is unique to small molecule TKI therapy and may facilitate the molecular switch.

involve intracellular signaling relationships and are well supported NO levels63,64 ! HTN63,64 ! atherosclerosis, whereas arm two
in the literature,5,10,11,63,64,69 are extracted from Figure 5 and is TKI therapy ! mitochondrial dysfunction11,63,64 ! endothe-
illustrated in isolation in Figure 6 for further clarity. In Figure 5, lial dysfunction (mediated by increased ROS levels, decreased
arm one of the rst feedforward loop is as follows. TKI therapy calcium levels, and decreased NO levels)63,64 ! atherosclerosis.
inhibits the PDGFR (among other multiple kinases). Interference In Figure 5 and Figure 6a, arm one of the third feedforward loop
with the PDGFR has been shown to lead to mitochondrial dys- is TKI therapy ! mitochondrial dysfunction11,63,64 ! decreased
function.11 Mitochondrial dysfunction results in decreased NO NO levels63,64 ! HTN63,64 ! atherosclerosis, whereas arm two is
levels,63,64 which leads to HTN.63,64 In arm two of this rst feed- TKI therapy ! decreased angiogenesis5,10 ! HTN63,64 ! athero-
forward loop, mitochondrial dysfunction reduces intracellular cal- sclerosis. In Figure 5 and Figure 6b, arm one of the fourth feedfor-
cium concentration, which also induces decreased NO levels and ward loop is TKI therapy ! mitochondrial dysfunction11,63,64 !
aggravates HTN,63,64 and risk for atherosclerosis. This could be endothelial dysfunction (mediated by increased ROS levels,
written more succinctly as: TKI therapy ! mitochondrial dys- decreased calcium levels, and decreased NO levels)63,64 ! athero-
function11,63,64 ! decreased NO levels63,64 ! HTN63,64 ! ath- sclerosis, whereas arm two is TKI therapy ! decreased angiogene-
erosclerosis, whereas arm two is TKI therapy ! mitochondrial sis5,10 ! HTN63,64 ! atherosclerosis.
dysfunction11,63,64 ! reduced calcium concentration63,64 ! In Figure 5, arm one of the fth feedforward loop is TKI
decreased NO levels63,64 ! HTN63,64 ! atherosclerosis. Similar- therapy ! mitochondrial dysfunction11,63,64 ! VSMC
ly, Figure 5 shows arm one of the second feedforward loop as dysfunction (mediated by increased ROS levels)63,64,69 ! athero-
TKI therapy ! mitochondrial dysfunction11,63,64 ! decreased sclerosis, whereas arm two is TKI therapy ! mitochondrial

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 75


Figure 6 One feedback and five feedforward loops in small molecule tyrosine kinase inhibitor (TKI) cardio-oncology. Several positive feedforward loops
(1FFLs) and one positive feedback loop (1FBL) are isolated here for visual clarification. Each positive feedforward loop has two arms. (a) Arm one of the
third feedforward loop (1FFL3) is TKI therapy ! mitochondrial dysfunction ! decreased nitric oxide (NO) levels ! hypertension (HTN) ! atherosclerosis,
whereas arm two is TKI therapy ! decreased angiogenesis ! HTN ! atherosclerosis. (b) Arm one of the fourth feedforward loop (1FFL4) is TKI therapy !
mitochondrial dysfunction ! endothelial dysfunction (mediated by increased reactive oxygen species (ROS) levels, decreased calcium levels, and
decreased NO levels) ! atherosclerosis, whereas arm two is TKI therapy ! decreased angiogenesis ! HTN ! atherosclerosis. (c) Arm one of the sev-
enth feedforward loop (1FFL7) is TKI therapy ! mitochondrial dysfunction ! vascular smooth muscle cell (VSMC) dysfunction (mediated by increased
ROS levels) ! atherosclerosis, whereas arm two is TKI therapy ! decreased angiogenesis ! HTN ! atherosclerosis. (d) Arm one of the eighth feedfor-
ward loop (1FFL8) is TKI therapy ! mitochondrial dysfunction ! adhesion/activation of inflammatory cells (mediated by increased ROS levels) ! athero-
sclerosis, whereas arm two is TKI therapy ! mitochondrial dysfunction ! decreased NO levels ! HTN ! atherosclerosis. (e) Arm one of the 10th
feedforward loop is (1FFL10) TKI therapy ! mitochondrial dysfunction ! adhesion/activation of inflammatory cells (mediated by increased ROS levels) !
atherosclerosis, whereas arm two is TKI therapy ! decreased angiogenesis ! HTN ! atherosclerosis. (f) The first feedback loop (1FBL1) is composed of
mitochondrial dysfunction and increased ROS levels.
dysfunction11,63,64 ! decreased NO levels63,64 ! HTN63,64 ! (Figure 3). Studies have shown that TKIs can inhibit these
atherosclerosis. In Figure 5, arm one of the sixth feedforward pumps.36,72,73 In fact, there is potentially a bell-shaped curve dis-
loop is TKI therapy ! mitochondrial dysfunction11,63,64 ! tribution of the ability of several TKIs at different concentrations
VSMC dysfunction (mediated by increased ROS levels)63,64,69 ! to activate or inhibit these efux pumps.72 Thus, inhibition or
atherosclerosis, whereas arm two is TKI therapy ! mitochondrial activation of efux pumps by small molecule TKIs can contribute
dysfunction11,63,64 ! endothelial dysfunction (mediated by to homeostasis within cardiac or vascular cells, while at the same
increased ROS levels, decreased calcium levels, and decreased NO time having the capacity to worsen toxicity. For example, inhibi-
levels)63,64 ! atherosclerosis. In Figure 5 and Figure 6c, arm one tion of doxorubicin efux pumps by small molecule TKIs can
of the seventh feedforward loop is TKI therapy ! mitochondrial increase intracellular doxorubicin levels, which can worsen double-
dysfunction11,63,64 ! VSMC dysfunction (mediated by increased strand breaks and ROS and subsequently apoptosis, as well as ath-
ROS levels)63,64,69 ! atherosclerosis, whereas arm two is TKI ther- erosclerosis. In this way, small molecule TKIs inhibiting efux
apy ! decreased angiogenesis5,10 ! HTN63,64 ! atherosclerosis. pumps adds another arm to the feedforward loops that instigate
Figure 5 and Figure 6d show arm one of the eighth feedforward the molecular switch. In fact, a number of small molecule TKIs
loop as TKI therapy ! mitochondrial dysfunction11,63,64 ! adhe- (e.g., cediranib, lapatinib, and sunitinib) have been used to inhibit
sion/activation of inammatory cells (mediated by increased ROS ABC transporters and efux pumps and thereby reverse drug
levels)63,64 ! atherosclerosis, whereas arm two is TKI therapy ! resistance.36 Although this improves outcomes and efcacy, this
mitochondrial dysfunction11,63,64 ! decreased NO levels63,64 ! methodology could also worsen toxicity. A ne balance would
HTN63,64 ! atherosclerosis. In Figure 5, arm one of the ninth need to be achieved between efcacy and toxicitythe same
feedforward loop is TKI therapy ! mitochondrial dysfunc- desired balance that drives the eld of cardio-oncology.
tion11,63,64 ! adhesion/activation of inammatory cells (mediated
by increased ROS levels)63,64 ! atherosclerosis, whereas arm two is IMPLEMENTATION OF SYSTEMS MEDICINE IN
TKI therapy ! mitochondrial dysfunction11,63,64 ! endothelial CLINICAL PRACTICE
dysfunction (mediated by increased ROS levels, decreased calcium Integrating various systems-based approaches into clinical prac-
levels, and decreased NO levels)63,64 ! atherosclerosis. In Figure 5 tice will likely provide the most meaningful outcomes for preci-
and Figure 6e, arm one of the tenth feedforward loop is TKI thera- sion medicine in cardio-oncology (Figure 2). High-powered
py ! mitochondrial dysfunction11,63,64 ! adhesion/activation of computers should be used to analyze the plethora of data, or as
inammatory cells (mediated by increased ROS levels)63,64 ! ath- commonly referred to big data, resulting from distinct systems-
erosclerosis, whereas arm two is TKI therapy ! decreased angio- based approaches. Computational and mathematical modeling
genesis5,10 ! HTN63,64 ! atherosclerosis. should be leveraged to assess combinatorial interactions among
In Figure 5 and Figure 6f, the rst feedback loop is composed various components of the cardiovascular system in response to
of mitochondrial dysfunction and increased ROS levels.63,64 In perturbation by TKIs. Such interactions often lead to complex
Figure 5, the second feedback loop is composed of mitochondrial
phenotypes that are not additives, but rather can be synergistic or
dysfunction and decreased calcium levels.63,64 Both loops would
antagonistic. Predictive computational modeling could fuse data
worsen mitochondrial dysfunction and ultimately atherosclero-
from the available systems medicine tools to determine actionable
sis.63,64 Both feedback loops are likely common to small molecule
feedback for implementation in precision medicine. The output
TKIs, monoclonal RTK antibodies, and anthracyclines, as all
should be combed through to help determine which individual
three can target pathways that induce mitochondrial dysfunction
patients maybe at high vs. low risk for TKI-induced cardiovascu-
and resultant downstream effects.4,11,43,53 For the same reason, of
lar toxicity. At the same time, data should be evaluated to also
the feedforward loops, six should be common to small molecule
determine which individuals might be considered responders or
TKIs, monoclonal RTK antibodies, and anthracyclines. Four
would be unique to small molecule TKI-induced atherosclerosis nonresponders to cardioprotective therapy.
the loops that include decreased angiogenesis. This has the The predictive P*4 pathway would be useful to streamline sys-
potential to produce a molecular switch, in which TKI therapy tems medicine in clinical practice.59 The pathway includes sys-
switches on atherosclerosis (in some patients), and in the absence tems medicine counseling, akin to genetic counseling, prior to
of the four unique feedforward loops involving decreased angio- systems medicine testing. A composite systems medicine score
genesis, the switch is off. This systems biology phenomenon may would categorize individuals as high-risk, low-risk, or potentially
help to explain why small molecule TKIs, relative to monoclonal intermediate-risk for developing TKI-induced cardiovascular tox-
RTK antibodies and anthracyclines, may show more of an associ- icity. Adjustments in cancer therapy and initiation of cardiopro-
ation with atherosclerosis and cardiovascular disease. This pro- tective therapy would be guided by the composite systems
vides a great example of how systems biology and likely network medicine score. Based on the progress of studies on cardiovascular
biology can aid in the interpretation of clinical or phenotypic toxicity from small molecule TKI therapy, the most specic
observations. approach with robust data is currently genomics (Figure 2, top
left). Clinical trials should be designed to determine whether
Influence of small molecule tyrosine kinase inhibitors genomic testing prior to small molecule TKI therapy could
on efflux pumps decrease the incidence of cardiotoxicity. This would be facilitated
Small TKIs also interact with doxorubicin therapy via ATP- by initiation of cardioprotective drugs for those deemed to be at
binding cassette transporters or efux pumps, such as MDR1 high risk (Figure 2, right), as well as minimizing other risk

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 77


factors or concomitant cardiotoxic therapy, in order to avoid Such drugs will alleviate injury to the heart, while facilitating
multiple hits (Figure 4). As the eld of cardio-oncology pro- cancer remission or cure.
gresses and the state of the art advances, iterative clinical trials
could be pursued to rene a composite systems medicine score, as
comprehensive data from additional systems-based approaches ACKNOWLEDGMENTS
become available. The Joerg Herrmann was supported by NIH grant HL 116952.

CONCLUSION C 2016 American Society for Clinical Pharmacology and Therapeutics


V
Opportunities for systems-based approaches to precision medi-
cine have been described, using the example of small molecule 1. Hurwitz, H. et al. Bevacizumab plus irinotecan, fluorouracil, and
TKIs in cardio-oncology. Primarily, ndings were presented from leucovorin for metastatic colorectal cancer. N. Engl. J. Med. 350,
genomic studies that have revealed variants associated with 23352342 (2004).
2. Lenneman, C.G. & Sawyer, D.B. Cardio-oncology: an update on
sunitinib-induced cardiovascular toxicity. These variants affect cardiotoxicity of cancer-related treatment. Circ. Res. 118, 1008
angiogenesis and TKI pharmacokinetics, metabolism, regulation, 1020 (2016).
transport, and efux. The potential for useful inferences from 3. Armenian, S.H. et al. Cardiovascular disease among survivors of
adult-onset cancer: a community-based retrospective cohort study. J.
other systems biology tools was also described. Such tools includ- Clin. Oncol. 34, 11221130 (2016).
ed transcriptomics, epigenomics, miRNA regulomics, proteomics, 4. Bellinger, A.M. et al. Cardio-oncology: how new targeted cancer
metabolomics, microbiomics, interactomics, and network biology therapies and precision medicine can inform cardiovascular
discovery. Ciculation 132, 22482258 (2015).
or medicine. Mathematical and computational biology or medi- 5. Krause, D.S. & Van Etten, R.A. Tyrosine kinases as targets for cancer
cine could also be used to model interactions among various car- therapy. N. Engl. J. Med. 353, 172187 (2005).
diovascular system components implicated in TKI-induced 6. Wilhelm, S. et al. Discovery and development of sorafenib: a
multikinase inhibitor for treating cancer. Nat. Rev. Drug Discov. 5,
toxicity. Robust predictive models spanning multiple scales 835844 (2006).
(e.g., genomics, transcriptomics, and proteomics) would augment 7. Herrmann, J. Tyrosine kinase inhibitors and vascular toxicity: impetus
clinical information available to physicians and scientists. The for a classification system? Curr. Oncol. Rep. 18, 33 (2016).
8. Carmeliet, P. Angiogenesis in life, disease and medicine. Nature
functional and phenotypic corollaries of variations in omics 438, 932936 (2005).
induced by TKI therapy could be integrated in predictive compu- 9. Bhargava, P. VEGF kinase inhibitors: how do they cause hypertension?
tational models to inuence patient care and outcomes. Further, Am. J. Physiol. Regul. Integr. Comp. Physiol. 297, R1R5 (2009).
10. Ghatalia, P. et al. Congestive heart failure with vascular endothelial
synergic toxicity likely occur when TKIs are used with anthracy- growth factor receptor tyrosine kinase inhibitors. Crit. Rev. Oncol.
clines or monoclonal antibodies to RTKs, with consolidation of Hematol. 94, 228237 (2015).
a myriad of feedback and feedforward loops and a molecular 11. Mercurio, V. et al. Models of heart failure based on the cardiotoxicity
of anticancer drugs. J. Card. Fail. 22, 449458 (2016).
switch. Our understanding of small molecule TKI intracellular 12. Kerkela, R. et al. Sunitinib-induced cardiotoxicity is mediated by off-
signaling and how this overlaps with other therapeutic pathways target inhibition of AMP-activated protein kinase. Clin. Transl. Sci. 2,
will be continually shaped by results from systems biology tools 1525 (2009).
13. Chintalgattu, V. et al. Cardiomyocyte PDGFR-beta signaling is an
in the future and knowledge of interaction with other therapeu- essential component of the mouse cardiac response to load-induced
tics, such as anthracyclines and HER2 receptor antibodies. The stress. J. Clin. Invest. 120, 472484 (2010).
potentially bell-shaped curve for activation and inhibition of 14. Herrmann, J. et al. Vascular toxicities of cancer therapies: the old and
the new--an evolving avenue. Circulation 133, 12721289 (2016).
MDR1 by numerous TKIs at dissimilar concentrations suggests 15. Vaziri, S.A., Kim, J., Ganapathi, M.K. & Ganapathi, R. Vascular
that several TKIs will impact ATP-binding cassette efux pumps endothelial growth factor polymorphisms: role in response and toxicity
differently. Although some individuals with no risk factors of tyrosine kinase inhibitors. Curr. Oncol. Rep. 12, 102108 (2010).
16. Garcia-Donas, J. et al. Single nucleotide polymorphism associations
will experience premature coronary heart disease, others develop with response and toxic effects in patients with advanced renal-cell
cardiovascular toxicity in the setting of underlying risk factors. carcinoma treated with first-line sunitinib: a multicentre, observational,
Contribution of multiple factors can lead to mitochondrial prospective study. Lancet Oncol. 12, 11431150 (2011).
17. Yuasa, T., Takahashi, S., Hatake, K., Yonese, J. & Fukui, I.
dysfunction and ROS in the vasculature, resulting in athero- Biomarkers to predict response to sunitinib therapy and prognosis in
sclerosis. Thus, cardiovascular toxicity occurs from multiple metastatic renal cell cancer. Cancer Sci. 102, 19491957 (2011).
hits at multiple levels, involving multiple cells, pathways, and 18. Mizuno, T. et al. ABCG2 421C>A polymorphism and high exposure of
sunitinib in a patient with renal cell carcinoma. Ann. Oncol. 21,
injuries. A similar systems biology approach will also be 13821383 (2010).
extremely valuable to dene the mechanism of accelerated ath- 19. van Erp, N.P. et al. Pharmacogenetic pathway analysis for determination
erosclerosis with the BCR-Abl inhibitors nilotinib and ponati- of sunitinib-induced toxicity. J. Clin. Oncol. 27, 44064412 (2009).
20. Junker, K., Ficarra, V., Kwon, E.D., Leibovich, B.C., Thompson, R.H. &
nib, which has remained, in large part, unexplained so far.7 To Oosterwijk, E. Potential role of genetic markers in the management of
minimize the impact of small molecule TKI therapy on the kidney cancer. Eur. Urol. 63, 333340 (2013).
cardiovascular system, the ABCDE steps are an easy approach 21. van der Veldt, A.A. et al. Genetic polymorphisms associated with a
prolonged progression-free survival in patients with metastatic renal cell
to remember for heart and vascular wellness of individuals cancer treated with sunitinib. Clin. Cancer Res. 17, 620629 (2011).
with cancer. Cardio-oncologic care can be further individual- 22. Diekstra, M.H. et al. Association analysis of genetic polymorphisms
ized by pursuing novel targets for drug development from in genes related to sunitinib pharmacokinetics, specifically clearance
of sunitinib and SU12662. Clin. Pharmacol. Ther. 96, 8189 (2014).
molecules implicated in vascular endothelial metabolism and 23. Kim, J.J. et al. Association of VEGF and VEGFR2 single nucleotide
signaling pathways elucidated in systems medicine studies. polymorphisms with hypertension and clinical outcome in metastatic

78 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


clear cell renal cell carcinoma patients treated with sunitinib. Cancer patients to doxorubicin-induced cardiotoxicity. Nat. Med. 22, 547
118, 19461954 (2012). 556 (2016).
24. Diekstra, M.H. et al. Association of single nucleotide polymorphisms 44. Xu, Q. et al. Micro-RNA-34a contributes to the impaired function of
in IL8 and IL13 with sunitinib-induced toxicity in patients with bone marrow-derived mononuclear cells from patients with
metastatic renal cell carcinoma. Eur. J. Clin. Pharmacol. 71, cardiovascular disease. J. Am. Coll. Cardiol. 59, 21072117 (2012).
14771484 (2015). 45. Tang, Y., Poteh, N.A., Chiang, A. & Kim, I. MicroRNA therapeutics for
25. Hacking, D. et al. Increased in vivo transcription of an IL-8 haplotype cardiac disease. Cardiovasc. Pharmacol. 2, 1000e113 (2013).
associated with respiratory syncytial virus disease-susceptibility. 46. De Rosa, S., Curcio, A. & Indolfi, C. Emerging role of microRNAs in
Genes Immun. 5, 274282 (2004). cardiovascular diseases. Circ. J. 78, 567575 (2014).
26. Petreaca, M.L., Yao, M., Liu, Y., Defea, K. & Martins-Green, M. 47. Pal, S.K. et al. Stool bacteriomic profiling in patients with metastatic
Transactivation of vascular endothelial growth factor receptor-2 by renal cell carcinoma receiving vascular endothelial growth factor-
interleukin-8 (IL-8/CXCL8) is required for IL-8/CXCL8-induced tyrosine kinase inhibitors. Clin. Cancer Res. 21, 52865293 (2015).
endothelial permeability. Mol. Biol. Cell. 18, 50145023 (2007). 48. Peng, X., Pentassuglia, L. & Sawyer, D.B. Emerging anticancer
27. Martin, D., Galisteo, R. & Gutkind, J.S. CXCL8/IL8 stimulates therapeutic targets and the cardiovascular system: is there cause for
vascular endothelial growth factor (VEGF) expression and the concern? Circ. Res. 106, 10221034 (2010).
autocrine activation of VEGFR2 in endothelial cells by activating 49. Burkard, T.R., Rix, U., Breitwieser, F.P., Superti-Furga, G. & Colinge, J.
NFkappaB through the CBM (Carma3/Bcl10/Malt1) complex. J. Biol. A computational approach to analyze the mechanism of action of the
Chem. 284, 60386042 (2009). kinase inhibitor bafetinib. PLoS Comput. Biol. 6, e1001001 (2010).
28. Eechoute, K. et al. Polymorphisms in endothelial nitric oxide synthase 50. Buchwald, F., Richter, L. & Kramer, S. Predicting a small molecule-
(eNOS) and vascular endothelial growth factor (VEGF) predict kinase interaction map: a machine learning approach. J. Cheminform.
sunitinib-induced hypertension. Clin. Pharmacol. Ther. 92, 503510 3, 22 (2011).
51. Kim, M., Gillies, R.J. & Rejniak, K.A. Current advances in
(2012).
mathematical modeling of anti-cancer drug penetration into tumor
29. Schneider, B.P. et al. Association of vascular endothelial growth
tissues. Front. Oncol. 3, 278 (2013).
factor and vascular endothelial growth factor receptor-2 genetic
52. Lin, F., Li, Z., Hua, Y. & Lim, Y.P. Proteomic profiling predicts drug
polymorphisms with outcome in a trial of paclitaxel compared with
response to novel targeted anticancer therapeutics. Expert Rev.
paclitaxel plus bevacizumab in advanced breast cancer: ECOG 2100.
Proteomics 13, 411420 (2016).
J. Clin. Oncol. 26, 46724678 (2008).
53. Hudis, C.A. Trastuzumab mechanism of action and use in clinical
30. Pander, J., Gelderblom, H. & Guchelaar, H.J. Pharmacogenetics of practice. N. Engl. J. Med. 357, 3951 (2007).
EGFR and VEGF inhibition. Drug Discov. Today 12, 10541060 54. Onitilo, A.A., Engel, J.M. & Stankowski, R.V. Cardiovascular toxicity
(2007). associated with adjuvant trastuzumab therapy: prevalence, patient
31. Bastide, A. et al. An upstream open reading frame within an IRES characteristics, and risk factors. Ther. Adv. Drug Saf. 5, 154166
controls expression of a specific VEGF-A isoform. Nucleic Acids Res. (2014).
36, 24342445 (2008). 55. Pentassuglia, L. & Sawyer, D.B. The role of neuregulin-1b/ErbB
32. Hood, J.D., Meininger, C.J., Ziche, M. & Granger, H.J. VEGF signaling in the heart. Exp. Cell Res. 315, 627637 (2009).
upregulates ecNOS message, protein, and NO production in human 56. Sysa-Shah, P. et al. Bidirectional cross-regulation between ErbB2 and
endothelial cells. Am. J. Physiol. 274(3 Pt 2), H1054H1058 (1998). b-adrenergic signalling pathways. Cardiovasc. Res. 109, 358373
33. Horowitz, J.R. et al. Vascular endothelial growth factor/vascular (2016).
permeability factor produces nitric oxide-dependent hypotension. 57. Abou-Alfa, G.K. et al. Doxorubicin plus sorafenib vs doxorubicin alone
Evidence for a maintenance role in quiescent adult endothelium. in patients with advanced hepatocellular carcinoma: a randomized
Arterioscler. Thromb. Vasc. Biol. 17, 27932800 (1997). trial. JAMA 304, 21542160 (2010).
34. Facemire, C.S., Nixon, A.B., Griffiths, R., Hurwitz, H. & Coffman, T.M. 58. Abou-Alfa, G. et al. Phase III randomized study of sorafenib plus
Vascular endothelial growth factor receptor 2 controls blood pressure doxorubicin versus sorafenib in patients with advanced
by regulating nitric oxide synthase expression. Hypertension 54, hepatocellular carcinoma (HCC): CALGB 80802 (Alliance). J. Clin.
652658 (2009). Oncol. 34(suppl. 4S), abstract 192 (2016).
35. Villanueva, C. & Giulivi, C. Subcellular and cellular locations of nitric 59. Brown, S.A., Sandhu, N. & Herrmann, J. Systems biology approaches
oxide synthase isoforms as determinants of health and disease. Free to adverse drug effects: the example of cardio-oncology. Nat. Rev.
Radic. Biol. Med. 49, 307316 (2010). Clin. Oncol. 12, 718731 (2015).
36. Sierra, J.R., Cepero, V. & Giordano, S. Molecular mechanisms of 60. Jones, L.W., Haykowsky, M.J., Swartz, J.J., Douglas, P.S. & Mackey,
acquired resistance to tyrosine kinase targeted therapy. Mol. Cancer J.R. Early breast cancer therapy and cardiovascular injury. J. Am. Coll.
9, 75 (2010). Cardiol. 50, 14351441 (2007).
37. Diekstra, M.H., Swen, J.J., Gelderblom, H. & Guchelaar, H.J. A 61. Herrmann, J. et al. Chronic proteasome inhibition contributes to
decade of pharmacogenomics research on tyrosine kinase inhibitors coronary atherosclerosis. Circ. Res. 101, 865874 (2007).
in metastatic renal cell cancer: a systematic review. Expert Rev. Mol. 62. Chintalgattu, V. et al. Coronary microvascular pericytes are the
Diagn. 16, 605618 (2016). cellular target of sunitinib malate-induced cardiotoxicity. Sci. Transl.
38. Shell, S.A., Lyass, L., Trusk, P.B., Pry, K.J., Wappel, R.L. & Bacus, Med. 5, 187ra69 (2013).
S.S. Activation of AMPK is necessary for killing cancer cells and 63. Tang, X., Luo, Y.X., Chen, H.Z. & Liu, D.P. Mitochondria, endothelial
sparing cardiac cells. Cell Cycle 7, 17691775 (2008). cell function, and vascular diseases. Front. Physiol. 5, 175 (2014).
39. Scott, J.M., Lakoski, S., Mackey, J.R., Douglas, P.S., Haykowsky, 64. Hulsmans, M., Van Dooren, E. & Holvoet, P. Mitochondrial reactive
M.J. & Jones, L.W. The potential role of aerobic exercise to modulate oxygen species and risk of atherosclerosis. Curr. Atheroscler. Rep.
cardiotoxicity of molecularly targeted cancer therapeutics. Oncologist 14, 264276 (2012).
18, 221231 (2013). 65. Ross, R., Faggiotto, A. Bowen-Pope, D. & Raines, E. The role of
40. Mirza, Z. et al. Molecular interaction of a kinase inhibitor midostaurin endothelial injury and platelet and macrophage interactions in
with anticancer drug targets, S100A8 and EGFR: transcriptional atherosclerosis. Circulation 70(5 Pt 2), 1117711182 (1984).
profiling and molecular docking study for kidney cancer therapeutics. 66. Weintraub, N.L., Jones, W.K. & Manka, D. Understanding
PLoS One 10, e0119765 (2015). radiation-induced vascular disease. J. Am. Coll. Cardiol. 55,
41. Eldridge, S., Guo, L., Mussio, J., Furniss, M., Hamre, J. & Davis, M. 12371239 (2010).
Examining the protective role of ErbB2 modulation in human-induced 67. Darby, S.C. et al. Risk of ischemic heart disease in women after
pluripotent stem cell-derived cardiomyocytes. Toxicol. Sci. 141, 547 radiotherapy for breast cancer. N. Engl. J. Med. 368, 987998 (2013).
559 (2014). 68. Bhatia, N. et al. Cardiovascular effects of androgen deprivation
42. Force, T. & Kolaja, K.L. Cardiotoxicity of kinase inhibitors: the therapy for the treatment of prostate cancer: ABCDE steps to reduce
prediction and translation of preclinical models to clinical outcomes. cardiovascular disease in patients with prostate cancer. Circulation
Nat. Rev. Drug Discov. 10, 111126 (2011). 133, 537541 (2016).
43. Burridge, P.W. et al. Human induced pluripotent stem cell-derived 69. Ku, D.D., Zaleski, J.K., Liu, S. & Brock, T.A. Vascular
cardiomyocytes recapitulate the predilection of breast cancer endothelial growth factor induces EDRF-dependent relaxation in

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 79


coronary arteries. Am. J. Physiol. 265(2 Pt 2), H586H592 radi, A., Sarkadi, B. & Ke
72. Hegedus, T., Orfi, L., Seprodi, A., Va ri, G.
(1993). Interaction of tyrosine kinase inhibitors with the human multidrug
70. Liu, Y., Li, H., Bubolz, A.H., Zhang, D.X. & Gutterman, D.D. transporter proteins, MDR1 and MRP1. Biochim. Biophys. Acta
Endothelial cytoskeletal elements are critical for flow-mediated 1587, 318325 (2002).
dilation in human coronary arterioles. Med. Biol. Eng. Comput. 46, 73. Wang, Y.J., Zhang, Y.K., Kathawala, R.J. & Chen, Z.S. Repositioning
469478 (2008). of tyrosine kinase inhibitors as antagonists of ATP-binding cassette
71. Madamanchi, N.R. & Runge, M.S. Mitochondrial dysfunction in transporters in anticancer drug resistance. Cancers (Basel) 6,
atherosclerosis. Circ. Res. 100, 460473 (2007). 19251952 (2014).

80 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt

Das könnte Ihnen auch gefallen