You are on page 1of 12

Cardio-Oncology: An Integrated

Discipline or Just a Portmanteau?*


Amit Varma, M.D., PhD, FACC
The Heart Group of Lancaster General Health
Advanced Heart Failure & Mechanical Circulatory Support
Congestive Heart Failure & Cardio-Oncology Clinics

INTRODUCTION artery disease (CAD) is second only to recurrence of


Cardio-oncology is a rapidly growing and evolving cancer as a leading cause of death in cancer survivors.
field that aims to optimize the cardiovascular (CV) care Cardio-oncology programs are thus essential to
received by cancer patients before and after chemo- identify risk factors for cardiotoxicity, to treat and
therapy. There are now more than 14.5 million cancer reduce them, and to monitor for CV toxicities from
survivors in the United States, and it is estimated that prior and ongoing chemo- and radio-therapy regimens.
by the year 2020, there will be roughly 18 million.1 The goal is to optimize cancer patients prior to chemo-
There is a broad range of cardiotoxic side effects therapy, to help identify cardiotoxicities early, and to
of chemotherapy and radiation treatment (Fig. 1). institute pharmacotherapy when necessary, in hopes of
Cardiotoxicity used to be considered just a reduction reducing morbidity and mortality from cardiac events
in resting left ventricular ejection function (LVEF), but without impeding or disrupting oncology treatment
it is now understood to include direct effects on cardiac regimens.
structure and function, the cardiac conduction system,
the systemic and pulmonary vasculatures, hemostasis THE BASELINE CLINICAL ASSESSMENT
and thrombosis, as well as direct injury and stress to In the cardio-oncology clinic, the risk of an
the endothelium. As a result of the latter, coronary individual patient developing cardiotoxicity from

Fig. 1. Cardiotoxic Side Effects of Chemotherapy and Radiation Treatment

Source: Lenneman CG, Sawyer DB. Cardio-Oncology: An update on Cardiotoxicity of Cancer-Related Treatment. Circ Res 2006;118: 1008-1020.

* pôrt manto – noun – a word blending the sounds and combining the meanings of two others, for example motel (from “motor” and
“hotel”) or brunch (from “breakfast” and “lunch”).

100 The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4
Cardio-Oncology

treatment is assessed well before chemotherapy begins. CARDIOTOXICITY


The two key factors are: 1) the specific chemotherapy Over the last four decades, strategies aimed at
and/or radiation therapy proposed, and 2) coexisting detecting cardiac dysfunction have evolved from inva-
CV disease, age and sex. Especially in older patients, sive, direct endomyocardial biopsies, to non-invasive
pre-existing conditions may limit therapeutic options, cardiac imaging. Terminology has also evolved, and the
so it is imperative prior to starting chemotherapy to term “Cancer Therapeutics Related (or Chemotherapy-
risk-stratify patients, to optimize blood pressure, lipids, Related) Cardiac Dysfunction (CTRCD) has been
and blood sugar, and to encourage tobacco cessation. adopted to describe LV dysfunction related to cancer
Patients found to have a subnormal LVEF should be treatment, as well as the type of myocardial injury.
started on goal-directed medical therapy for congestive CTCRD is defined as a > 5% drop in LVEF in
heart failure to optimize heart function. The cardi- symptomatic heart failure (HF) patients, or a >10%
ologist and oncologist should collaboratively consider drop in LVEF to <53% in the asymptomatic patient.
whether cardiotoxic chemotherapy is in the best inter- There are two main types of CTRCD:
est of the patient. Patients at highest risk may benefit a) Type I is dose-dependent, irreversible, and
from cardioprotective medical therapy with beta-block- results from ultra-structural changes within the myo-
ers, statins, angiotensin converting enzyme (ACE) cardium. It is typified by the anthracycline class of
inhibitors, or angiotensin receptor blockers (ARB).2 drugs which inhibit function of topoisomerase 2B
Interestingly, many of the risk factors associated within cardiomyocytes, leading to DNA double strand
with the development of CV disease, such as tobacco breaks, mitochondrial dysfunction, and development
use, are also risk factors for developing cancer. It is of reactive oxygen species that lead to damage or death
postulated that inflammation not only increases the of the cardiomyocytes.
risk of malignancy, but is the common denominator b) Type II is mostly reversible, is not dose-depen-
that links the development of CV diseases, diabetes, dent, and is not associated with ultra-structural
atherosclerosis, endothelial dysfunction, and meta- changes in the myocardium, so functional recovery is
bolic syndrome. High C-reactive protein (CRP) levels often seen after interrupting and discontinuing ther-
are associated with an increased incidence of CV apy. Trastuzumab (Herceptin®) is a typical causative
events and an increased risk of developing a malig- agent, but since it was approved, dozens of other che-
nancy, and a higher CRP portends worse survival.1 In motherapy agents have been introduced that also cause
regard to the interactions between various molecular reversible cardiotoxicity, especially the small-molecule
pathways that are not yet fully elucidated, it is relevant tyrosine kinase inhibitors (TKI) and vascular endothe-
that the same risk factors for coronary atherosclero- lial growth factor (VEGF) inhibitors. Their net effect
sis also increase the risk of CV complications from on the CV system is still being studied and the car-
chemotherapy. diotoxicity rates are unpredictable. Attempts have been
A baseline cardiac assessment should be per- made to propose a unified approach to managing these
formed on patients with the following cardiac risk complex patients, but there is no single, evidence-
factors: left ventricular (LV) dysfunction, age<18 or based, guideline for management, and the approach
>65, planned chemotherapy with multiple cardio- to treatment, evaluation, and drug stoppage is varied
toxic agents or high doses of a single cardiotoxic at best.
drugs, known prior cardiac pathology (LV hypertro- The incidence of cardiomyopathy, potential CV
phy, hypertension, CAD), diabetes mellitus or prior toxicities and current uses of common chemothera-
radiation exposure.1 Risks should be minimized and peutic agents are summarized in Table 1 (pages 102
patients should be treated prior to chemotherapy; as & 103).
feasible, patients may benefit from referral to a cardio-
oncologist who specializes in treating and following ANTHRACYCLINES
cancer patients through survivorship. Nevertheless, Cardiac dysfunction from cancer therapeutics
the large and growing number of cancer patients was first widely recognized in the 1960s with a new
makes it prudent for internists, generalists, cardiolo- class of chemotherapeutics—the anthracyclines – a
gists, oncologists, and hematologists to all become type of antibiotic derived from Streptomyces bacteria.
well versed in risk-stratifying and caring for these Though their use continues to be limited by cardiotox-
patients in regard to cardiotoxicity. icity, they are used for a variety of cancers including

The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4 101
102
Table 1. The Incidence of Cardiomyopathy, Potential CV Toxicities and Current Uses of Common Chemotherapeutic Agents

The Journal of Lancaster General Hospital



Winter 2017

Vol. 12 – No. 4
Cardio-Oncology
Table 1. The Incidence of Cardiomyopathy, Potential CV Toxicities and Current Uses of Common Chemotherapeutic Agents (continued)
Cardio-Oncology

The Journal of Lancaster General Hospital



Winter 2017

Vol. 12 – No. 4
103
Cardio-Oncology

lymphoma, leukemia, sarcoma and breast cancer, and of patients with tumors that overexpress the HER2
are still among the most effective chemotherapeutic protein, which tend to be rapidly progressive, but car-
agents.3 Exposure to these drugs causes both short- diotoxicity is an ever-present risk.
and long-term complications and risk largely depends In one meta-analysis, treatment with trastuzumab
on the cumulative dose. At a cumulative dose of 400 significantly increased the incidence of congestive
mg/m2, the risk of developing HF is around 5% but heart failure (RR 5.11, 90% CI [3.0-8.7]; P<0.000001)
increases dramatically to about 25% when the cumula- and the LVEF declined significantly (RR 1.83, 90%
tive dose reaches 700 mg/m2. 4 A thorough assessment CI [1.36-2.47], P=0.0008).8 In pre-clinical studies,
to identify underlying risk factors is essential, as patients cardiac dysfunction with HER2 targeted therapies
with hypertension, CAD, diabetes, LV dysfunction, or occurred secondary to disruption in signaling between
prior radiation are at much higher risk for developing the HER2 receptor and the ligand growth factor,
toxicity at much lower doses. These patients must be Neuregulin.2 The HER2 (ErbB2)-Neuregulin receptor
closely followed during and after therapy and the total ligand-signaling cascade is critical to myocyte growth,
cumulative dose must be appropriately adjusted, or the survival and homeostasis, and HER2 targeted thera-
drug may have to be changed.5,6 Acute cardiac dysfunc- pies interrupt myocyte homeostasis and cardiac repair
tion is reported in up to 3.2% of patients receiving by affecting the signaling pathway. Furthermore, this
anthracycline therapy and usually occurs within weeks signaling pathway regulates vasomotor tone and sym-
of starting therapy. These patients usually present with pathetic output within the CV system; breast cancer
arrhythmias, HF, LV dysfunction, and myocarditis-peri- patients treated with HER2 antagonists experience
carditis syndrome, but occult LV dysfunction may be increased levels of norepinephrine, heart rate and
present in up to 20% of patients, and may not be clini- blood pressure.2
cally evident until the completion of chemotherapy. Newer HER2 targeted therapies have emerged in
Late complications of anthracycline therapy include the last decade including lapatinib, an oral HER2 tar-
the development of HF decades after treatment. geted small molecule, dual TKI of the Her2/neu and
The use of an encapsulated (“pegylated” by a lipo- the epidermal growth factor receptors. Pertuzamab,
somal moiety) form of doxorubicin (AdriamicinTM) allows another HER2 targeted agent, was designed to over-
a higher cumulative dose with a lower rate of myocar- come trastuzumab resistance. Thus far, clinical studies
dial damage, but with similar efficacy. Despite this, with the newer HER2 targeted agents have not shown
however, its use does not eliminate the advisability of clear evidence for causing cardiotoxicity.1 Routine
routine surveillance of LV function during therapy. monitoring of cardiac function during treatment with
Another approach is the use of dexrazoxane, an any HER2 antagonist has become standard practice
iron chelator like EDTA that protects the myocardium and most expert panels, based on clinical studies, rec-
from the effects of doxorubicin or epirubicin. These ommend echocardiograms every three months during
cardioprotective effects have been clinically confirmed therapy.
in a number of studies, but there are concerns about The novel HER2 antagonists have significantly
a lower response rate and development of secondary improved outcomes in breast cancer patients, but we
leukemia in childhood survivors.7 Epirubicin is less must identify high-risk patients, frequently monitor
cardiotoxic than doxorubicin and in studies has been for the development of cardiotoxicity during and after
shown to decrease the risk of both clinical and subclin- therapy, and initiate HF therapies when appropriate.
ical cardiotoxicity.7 Nevertheless, it is still cardiotoxic
and the Food and Drug Administration has limited ALKYLATING AGENTS
the cumulative dose to < 900 mg/m2 per patient. Cisplatin is a commonly used alkylating agent in
several solid organ tumors (genitourinary, gastroin-
HER2 ANTAGONISTS testinal, head and neck), and vascular toxicity is the
HER2/neu antagonists, typified by trastuzumab primary concern. Given the high cure rates, long-term
(Herceptin®), are a relatively new type of chemother- consequences post treatment are emerging, particularly
apy for breast cancer – monoclonal antibodies that CV diseases, including hypertension, dyslipidemia,
target and inhibit the Human epidermal growth fac- early atherosclerosis, CAD, Raynaud’s phenomenon,
tor receptor-2 (also known as ErbB2). Trastuzumab has and thromboembolic events. Testicular cancer sur-
dramatically improved the formerly dismal prognosis vivors treated with Cisplatin-based therapy have an

104 The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4
Cardio-Oncology

increased risk of premature atherosclerosis, increased studies with sunitinib have shown that LV dysfunction
biomarkers suggestive of endothelial injury, and devel- and HF symptoms respond well to discontinuing the
opment of CAD and vascular disease.1 Cisplatin is offending drug and starting goal directed HF thera-
incompletely eliminated after treatment, and platinum pies. However, pre-clinical animal and cellular data
can be measured in the serum years after therapy, lead- show sunitinib can promote and induce cardiomyocyte
ing to speculation that this continuous small amount apoptosis thus at least some element of irreversible
of Cisplatin leads to endothelial dysfunction. cardiac damage does occur. Recent meta-analyses have
Cyclophosphamide has also been associated with shown cardiac dysfunction and HF to be a relatively
cardiotoxicity that is not clearly dose-dependent, common problem with TKIs, but the true incidence of
including reduced LVEF, decreased electrocardiogram TKI-induced cardiomyopathy with each TKI, and what
(ECG) voltage with or without pericardial effusions, the ideal screening and monitoring period is for these
and – most severely – hemorrhagic myopericarditis with patients, has yet to be determined.
pericardial effusion, attributed to endothelial capillary
damage. This last complication can occasionally lead MICROTUBULE INHIBITORS
to cardiac tamponade, but otherwise these scenarios The taxane family of chemotherapy agents
are managed conservatively. The risk of cardiotoxicity includes paclitaxel and docetaxel derived from the Taxus
is increased by prior radiation therapy to the chest and genus of plants. They exert their antineoplastic effect
mediastinum, older age, and reduced LVEF. by disrupting the function of microtubules, which are
vital for cell division. The taxanes have been used since
MULTI-TARGETED TKI AND VEGF INHIBITORS the 1990s to treat solid organ tumors including breast,
Angiogenesis is a requirement for adequate tumor ovarian, and non-small cell lung cancers. They stabilize
cell growth, and multiple anti-tumor therapies target guanosine diphosphate-bound tubulin in the microtu-
the angiogenesis-signaling cascade. The first of these bules, thereby inhibiting the process of cell division,
were inhibitors against VEGF and more recently small thus halting mitosis altogether.
molecules that inhibit VEGF receptor tyrosine kinase Arrhythmias are the most common side effect
and non-receptor tyrosine kinases. However, since with taxanes, including bradycardia and heart block,
VEGF plays an integral role in angiogenesis, endothe- suggesting the possibility that microtubules play a role
lial cell survival, vasodilatation, and cardiac contractile in calcium handling. It is speculated that paclitaxel
function, alterations in angiogenesis inevitably impact decreases the calcium amplitude and contraction of
CV biology and function. The small molecule recep- isolated cardiomyocytes, but shortens the time from
tor TKIs have an even more complex and variable CV maximum contracted state to relaxation. In general,
effect on the cardiac system than do the monoclonal bradyarrhythmias are asymptomatic and self-limited,
antibody based biologics. but supraventricular tachyarrhythmias such as atrial
VEGF also regulates endothelial cell health, fibrillation, atrial flutter, and atrial tachycardia have
survival, and repair; thus inhibition promotes microvas- all occurred. Docetaxel has been shown to potenti-
cular injury and potentiates thrombosis. In addition to ate the cardiotoxic effect of anthracycline therapy.9
causing hypertension and cardiac dysfunction, VEGF- Cardiotoxicity with the vinca alkalkaloids is rather
targeted therapies can promote thromboembolism uncommon, and may be more common with vinblastine
and are associated with a three-fold increase in arte- than with vincristine and vinorelbine. Symptoms usually
rial thromboembolic events such as transient ischemic include hypertension, myocardial ischemia, infarction,
attacks, strokes, angina, and myocardial infarction. and other vaso-occlusive complications.
VEGF signaling is also known to mediate adap-
tation of the myocardium to pressure overload. ANTIMETABOLITES
Suppressing VEGF during pressure overload leads to Antimetabolites are a class of chemotherapeutics
a rapid deterioration of heart function and promotes that interfere with DNA and RNA growth by substitut-
adverse cardiac remodeling. This phenomenon has ing the building blocks of DNA/RNA and damaging
been observed with bevacizumab (an anti-VEGF mono- the proliferating cells during the S phase of mitosis.
clonal antibody) and with sunitinib and a number of Commonly used antimetabolite drugs include: 5-fluo-
other multi-TKIs.1 It is still unclear whether cardiac rouricil (5-FU), cytarabine, gemcitabine, capecitabine,
dysfunction secondary to multi-TKIs is reversible. Early methotrexate, and hydroxyurea and are used to treat solid

The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4 105
Cardio-Oncology

tumors such as breast, ovarian, and gastrointestinal. TREATMENT OF CTRCD


Fluorouracil’s incidence of cardiotoxicity can approach (Chemotherapy-Related Cardiac Dysfunction)
20%. There are no formal guidelines from the American
The pathophysiology of cardiotoxicity is multifac- Heart Association/American College of Cardiology or
torial with several speculated mechanisms. Endothelial Heart Failure Society of America for the treatment
damage, thrombosis, and increased oxidative stress all and management of chemotherapy/radiation-induced
lead to cellular damage; coronary artery spasm leads to cardiotoxicity, but the European Society of Medical
myocardial ischemia and thus reduced oxygen delivery. Oncology recommends serial monitoring of cardiac
The most notable CV side effects include myocardial function and initiation of therapy with ACE inhibi-
ischemia, angina, and ECG changes including ST-T tors and beta-blockers in patients who develop LV
wave and T wave changes. dysfunction, or prophylactically in patients at high risk
Generally, cardiotoxicity occurs within the first of cardiotoxicity.11
few doses, and is much more likely to occur with These recommendations are based on several small
higher doses or continuous infusions. Currently there clinical studies. Recent prospective studies have looked
are no recommendations to pre-treat patients with at the cardioprotective role of ACE inhibitors and beta-
anti-anginal drugs, such as nitroglycerin or calcium blockers during chemotherapy (Fig. 2, next page). Early
channel blockers. However, some small trials have detection of cardiotoxicity provides an opportunity to
shown improvement in angina symptoms with 5-FU or prevent or reverse progression to advanced and end-
capecitabine when calcium channel blockers or nitrates stage HF, but there is still a paucity of data to guide
are used. Not surprisingly, the incidence of ischemia selection of surveillance intervals, imaging technolo-
related to 5-FU is much higher in patients with known gies, or biomarker tests, nor what thresholds should
underlying CAD.1 A subsequent re-challenge with trigger initiation of pharmacotherapy to prevent fur-
5-FU usually reproduces symptoms. Capecitabine is a ther toxicity.
prodrug that is metabolized to its active moiety, 5-FU, A key study showed that a critical factor in cardiac
and has cardiotoxicity similar to infused 5-FU. Patients recovery from anthracycline-induced cardiac dysfunc-
that have vasospasm induced by 5-FU will likely expe- tion is the elapsed time from the end of chemotherapy
rience recurrent symptoms with capecitabine and to the start of HF therapy with ACE inhibitors and
accordingly, management of cardiotoxicity is similar beta-blockers.6
to that with 5-FU. Fludarabine can cause chest pain The PRADA (Prevention of Cardiac Dysfunction
and hypotension, and the combination of fludara- During Adjuvant Breast Cancer Therapy) trial was a
bine with melphalan as a conditioning regimen prior randomized, placebo-controlled, double-blind study
to stem cell transplantation has also been associated to determine whether an angiotensin receptor blocker
with cardiac dysfunction.10 (candesartan), and/or a beta-blocker (metoprolol) may
prevent the development of LV dysfunction in patients
PROTEOSOME INHIBITORS receiving standard chemotherapy for early breast can-
The proteasome plays a key role in the main- cer.12 A total of 120 patients were randomized: LV
tenance of cardiac structure and function, and the ejection fraction declined 0.8% in the candesartan
ubiquitin-proteasome system regulates protein turn- group, versus 2.6% in the placebo group (P=0.026).
over in addition to activating cell-signaling pathways for The authors concluded that candesartan protected
growth and survival of tumors. Bortezomib (Velcade®), against a decline in LVEF, whereas metoprolol did not.
the first proteasome inhibitor available for clinical use, Many argued that this randomized study should have
suppresses plasma cell proliferation and is used to treat used carvedilol instead of metoprolol, but until recently
multiple myeloma. all studies in the cardio-oncology literature had been
Currently, there are only few reports of HF occur- retrospective in design and observational.
ring during treatment. Carfilzomib, one of the newer In another prospective trial (OVERCOME), 90
proteasome inhibitors, is an irreversible proteasome patients with acute leukemia undergoing anthracycline
inhibitor used for relapsed multiple myeloma. It has a chemotherapy were randomized to start enalapril and
much greater tendency to precipitate CV events such carvedilol versus placebo.13 At six months, prophylactic
as acute coronary syndrome, sudden cardiac death, enalapril and carvedilol stabilized LVEF compared with
and HF. placebo, but the difference of 3.1 by echocardiography

106 The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4
Cardio-Oncology

Fig. 2. The Cardioprotective Role of ACE Inhibitors and Beta-Blockers During Chemotherapy

Extensively adapted from Plana JC, Galderski M, Barac A, et al. Expert Consensus for Multimodality Imaging Evaluation of Adult Patients during
and after Cancer Therapy: A Report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging.
J Am Soc Echocardiogr 2014; 27: 911-939.

The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4 107
Cardio-Oncology

did not reach statistical significance (P=0.09). heart; and if concomitant antineoplastic agents are
In the MANTICORE study, which recently given, especially anthracyclines.15
reported five-year results, the ACE inhibitor perin- Long-term follow-up and evaluation should
dopril and the beta-blocker bisoprolol were evaluated include a yearly physical examination with a focused
in patients with HER2-positive early breast cancer assessment for signs and symptoms of peripheral
who received trastuzumab.14 The combination therapy vascular disease and RIHD. Asymptomatic patients
protected against declines in LVEF, with fewer inter- should receive a transthoracic echocardiogram five
ruptions in therapy. Secondary analyses of 33 patients years post exposure in high risk individuals and 10
who received perindopril, 31 patients who received years post exposure in all other patients. Similarly, a
bisoprolol, and 30 who received placebo for one year, cardiac stress test should be performed five years post
along with trastuzumab, showed that bisoprolol was more exposure in high risk patients and after 10 years in
effective than perindopril or placebo in preventing che- all others to assess for accelerated CAD secondary to
motherapy-mediated decline in LVEF: (–1% ± 5%) vs. radiation vasculopathy.15 Concurrent metabolic risk
(–3% ± 4%) and (–5% ± 5%) respectively (P = .001). factors such as hypertension, tobacco use, obesity,
However, cardiac MRI after 17 cycles of trastu- and diabetes, increase the incidence and severity of
zumab showed that indexed LVEDP (left ventricular cardiac disease.
end diastolic volume), increased in patients treated Early reports indicated that the proximity of the
with perindopril (+7 ± 14 mL/m2), bisoprolol (+8 mL ± 9 heart to the chest wall in breast cancer and Hodgkin’s
mL/m2), and placebo (+4 ± 11 mL/m2; P = .36). Thus, lymphoma increased the occurrence of RIHD, and
neither therapy prevented trastuzumab-mediated LV there were higher morbidity and mortality rates in
remodeling, as reflected in LVEDP as a surrogate for patients with left-sided breast cancer compared with
LV remodeling. The study was stopped early because right-sided cancer. However, later reports from the
this primary end point was not met, but the authors, 1993 Surveillance, Epidemiology, and End Results
and experts in the field, did conclude that identifi- (SEER) database demonstrated that there was no
cation of CV risk factors and optimization prior to difference in cardiac morbidity between left- and
initiation of cancer therapy is a key prevention strategy right-sided breast cancer treated with radiation ther-
for those at highest risk of developing cardiotoxicity. apy.16 After 1979, regardless of which breast, adding
It is anticipated that larger randomized clinical radiation to anthracycline treatment increased the
trials will help further delineate which HF therapies risk of valvular heart disease and HF, but not the risk
provide the greatest long-term benefits. In this regard, of myocardial infarction. This reduction in infarction
those active in the field of cardio-oncology strive to risk was attributed to a change in radiation technique
keep pace with the rapid evolution of chemothera- and delivery.17
peutics, and the incidence and consequences of The pathophysiology of RIHD is still not thor-
treatment-related CV side effects. oughly understood, but it is thought to be related
to fibrosis within the myocardium, pericardium,
RADIATION-INDUCED HEART DISEASE valves, and blood vessels. The cusps and leaflets of
Patients who receive radiation therapy are also the heart valves undergo fibrotic changes with and
at risk for long term CV toxicity. Radiation-induced without calcification, and for reasons still not under-
heart disease (RIHD) usually manifests 10-15 years stood, left-sided valves are more commonly affected
post exposure and can contribute to the develop- by radiation – which suggests that higher systemic
ment of: restrictive cardiomyopathy, valvular and pressures may play a role. Recent studies show a
pericardial disease, premature coronary artery dis- decline in RIHD in the past few decades, doubtless
ease, systolic and diastolic dysfunction, dysrhythmias, due to changes in delivery techniques, use of linear
autonomic dysfunction, and peripheral artery dis- accelerators, decreases in variation of doses at field
ease. Long term risk increases dramatically in patients edges, and use of computed tomography simulation
younger than 50 years of age; when the cumulative to help exclude the cardiac silhouette from tangential
dose of radiation exceeds 30 Gray (Gy) or the frac- beams.1 Nevertheless, we continue to treat patients
tional doses exceed 2 Gy per day; with radiation that were given radiotherapy during various time peri-
delivered to the anterior chest or left chest; with lack ods, especially childhood cancer survivors now in late
of adequate shielding; when the tumor is near the adulthood, and we must be aware of the spectrum of

108 The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4
Cardio-Oncology

vascular, myo-pericardial and valvular heart disease, the mechanism of toxicity caused by type II agents,
and aggressively treat modifiable risk factors. simply discontinuing the agent temporarily may be
sufficient to allow full myocardial recovery since
ASSESSMENT OF LEFT VENTRICULAR FUNCTION there is no irreversible damage to cardiomyocytes.
The European Society of Medical Oncology has Until unequivocal, evidence-based, clinical guide-
published algorithms for serial cardiac monitoring lines are available, our practice is to treat all patients
of patients at risk for type I and type II CTRCD. with reduced EF with HF therapies regardless of type
In 2014 the American Society of Echocardiography I or II CTRCD and to optimize medications to allow
with the European Association of Cardiovascular minimal interruptions to chemotherapy.
Imaging published a consensus statement on imag- When monitoring for CTRCD, one significant
ing assessment of patients during and after cancer limitation of echocardiography is that changes in
treatment.11,18 EF secondary to chemotherapy often occur when
All algorithms begin with a baseline assessment irreversible damage to cardiomyocytes has already
of LV function and use of echocardiography as the occurred. Since early initiation of medical therapy
modality of choice for serial assessment because of increases the likelihood of subsequent improvement
its wide spread availability and lack of radiation expo- in EF, determining the best methods to detect cardio-
sure. Radionucleotide angiography and cardiac MRI toxicity before it manifests as a decline in LVEF is an
can also be used, but the same modality should be area of intense and ongoing research.
utilized for all follow-up imaging. If initial assess- One technique that has shown promise is left
ment shows an abnormal LVEF, Cardio-oncology ventricular strain and strain rate imaging. Global
consultation should be sought for recommendations longitudinal strain (GLS) can be incorporated into
and possibly pharmacotherapy prior to initiation of routine echocardiography with readily available tech-
chemo- or radio-therapy. nology. For predicting declines in LVEF, subclinical
If the baseline LVEF is normal, then timing and LV dysfunction, or acute heart failure, several reviews
follow-up of subsequent EF assessment is based on have found GLS to have sensitivity ranging from
whether the cardiotoxic agent is associated with type 65-86% and specificity from 73-95%.19
I or type II CTRCD. (Fig. 2) If LV dysfunction is An Expert Consensus Panel concluded from
detected during chemotherapy, referral to a cardio- the available data that a reduction in GLS between
oncologist should be made promptly for immediate studies of less than 8% is not clinically relevant, a
initiation of ACEi/ARB and/or beta-blocker. reduction greater than 15% is a harbinger of subclini-
Time to initiation of therapy has been shown cal LV dysfunction, and a reduction of 8-15% is a gray
to be clinically relevant in patients receiving type I zone that warrants closer follow-up.18 At Lancaster
cardiotoxic agents. In patients experiencing type II General Health, we use the Phillips iE33 (Phillips
CTRCD, the offending agent can usually be reiniti- Medical System) ultrasound machine to perform two-
ated after a period of discontinuing the cardiotoxic and three- dimensional echocardiography and LV
drug. Whether use of medical therapy such as beta- GLS. Table 2 lists the normal values by gender and
blockers and/or ACE inhibitors/ARBs offer clinically age (divided into deciles), and as can be seen, they
significant improvements in outcomes in Type II car- can vary substantially.20
diotoxicity is still unknown; experts argue that given As noted earlier, it is not certain whether

The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4 109
Cardio-Oncology

treatment when subclinical LV dysfunction is first translated into no increases in cardiac events during
identified will prevent clinically significant HF or follow-up.
a loss of LV function; current data do not support It remains unclear whether optimal timing of
treating subclinical LV dysfunction. The Expert troponin assessment, or a single assessment, have
Consensus Panel advises a repeat echocardiogram prognostic implications. Currently, we do know that
within two to three weeks when a significant change patients with negative troponins during chemotherapy
in LVEF is noted on echocardiography. They also cau- (checked post cycle), have a lower risk of cardiac events
tion that when making major clinical decisions such or a drop in LV function than those with a positive
as discontinuing potentially life-saving chemotherapy, cTnI.1 Patients with a positive cTnI while receiving
it may be sensible to obtain cardiac MRI, either to chemotherapy should be referred to cardio-oncology
confirm a significant drop in LVEF, or if estimation for intense medical optimization, further risk strati-
of LV function by echocardiogram is unreliable or fication, and close follow-up and monitoring for LV
difficult to ascertain.18 dysfunction.

BIOMARKERS NT-proBNP
N-terminal prohormone of brain natriuretic
Cardiac troponins peptide (NT-proBNP) is another biomarker that has
Cardiac troponin I (cTnI) is the gold standard recently been evaluated for detection of early LV fail-
biomarker when diagnosing myocardial injury in che- ure in chemotherapy patients. Though studies are
motherapy patients, and is both sensitive and specific; ongoing, we still lack data to support its routine use.
in patients treated with anthracyclines, elevation in However, since acute elevations of NT-proBNP levels
cTnI identifies patients at risk for the development of raise concern for increased LV filling pressures, its
subsequent CTRCD.21 negative predictive value may be more useful, even
One large study of 703 patients measured cTnI though its variability in levels over time limits its util-
levels with each cycle of high dose anthracycline che- ity in heralding a decline in LV function.
motherapy, and 1 month after completion.6 The
majority of events occurred in cTnI positive patients, CONCLUSIONS
and a persistent increase in troponin was associated In the 21st century, as medicine has become
with an increase in the severity of CTRCD and a more subspecialized and – as a result – increasingly
higher incidence of cardiac events compared with only fragmented, it has become increasingly important
a transient increase. Troponin had a 99% negative to have close collaboration between the medical
predictive value, in that patients who had continually subspecialties. The rapidly emerging discipline of
negative troponin determinations remained at the low- Cardio-oncology takes a multidisciplinary approach,
est risk for CTRCD. A number of smaller studies have with alliances among cardiologists, oncologists, and
also demonstrated a correlation between troponin lev- hematologists. All work together for prevention,
els and LV dysfunction.22 early detection, and management of CV diseases in
Troponin may also be used to identify early car- cancer patients throughout all stages of treatment
diac injury in patients receiving treatment with HER2 and continuing into survivorship.
antagonists. The largest study conducted evaluated As newer antineoplastic agents are approved, the
251 patients receiving trastuzumab and found that cTnI magnitude of long-term CV morbidity is likely to
positivity was associated with an increased incidence of increase, with unknown long-term sequelae. Ideally,
cardiac events and lower likelihood of cardiac recovery. a close partnership and open discussion of patient
In recent studies, up to 11% of patients receiving management options between oncologists and cardi-
TKIs and VEGF inhibitors had elevated cTnI levels, ologists will ensure optimal care for patients receiving
but beta-blockers and aspirin normalized them, which cardiotoxic therapies.

110 The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4
Cardio-Oncology

References

1. Lenneman CG, Sawyer DB. Cardio-Oncology: An update on 13. Bosch X, Rovira M, Sitges M, et al. Enalapril and carvedilol for
Cardiotoxicity of Cancer-Related Treatment. Circ Res 2006;118: preventing chemotherapy-induced left ventricular systolic dysfunc-
1008-1020. tion in patients with malignant hemopathies: the OVERCOME
trial. J Am Coll Cardiol 2013; 61: 2355-2362.
2. Lenneman CG, Abdallah WM, Smith HM, et al. Sympathetic ner-
vous system alterations with HER2+ antagonism: an early marker 14. Pituskin E, Mackey JR, Koshman S, et al. Multidisciplinary
of cardiac dysfunction with breast cancer treatment? E Cancer Approach to Novel Therapies in Cardio-Oncology Research
Medical Science 2014; 8: 446. (MANTICORE 101-BREAST): A Randomized Trial for the
Prevention of Trastuzumab-Associated Cardiotoxicity. J Clin Oncol
3. Geisberg CA, Sawyer DB. Mechanisms of of anthracycline cardio-
2017; 35 : 870-877.
toxicity and strategies to decrease cardiac damage. Curr Hypertens
Rep. 2010; 12: 404-410. 15. Lancellotti P, Nkomo VT, Badano LP, J, et al. 2013 Expert
consensus for multi-modality imaging evaluation of cardiovas-
4. Shan K, Lincoff AM, Young JH. Anthracycline-induced cardiotox-
cular complications of radiotherapy in adults: a report from
icity. Ann Intern Med 1996;125: 47-58.
the European Association of Cardiovascular Imaging and the
5. Cardinale D, Colombo A, Torrisi R, et al. Trastuzumab-induced American Society of Echocardiography. J Am Soc Echocardiogr 2013;
cardiotoxicity: clinical and prognostic implication of troponin I 26: 1013-1032.
evaluation. J Clin Oncol 2010;28: 3910-3916.
16. Patt DA, Goodwin JS, Kuo YF, et al. Cardiac morbidity of adjuvant
6. Cardinale D, Sandri MT, Colombo A, et al. Prognostic value of tro- radiotherapy for breast cancer. J Clin Oncol 2005; 23: 7475-7482.
ponin I in cardiac risk stratification of cancer patients undergoing
17. Hooning MJ, Botma A, Aleman BM, et al. Long-term risk of car-
high dose chemotherapy. Circulation 2004;109: 2749-2754.
diovascular disease in 10-year survivors of breast cancer. J Natl
7. Smith LA, Cornelius VR, Plummer CJ, et al. Cardiotoxicity of Cancer Inst 2007; 99: 365-375.
anthracycline agents for the treatment of cancer: systematic review
18. Plana JC, Galderski M, Barac A, et al. Expert Consensus for
and meta-analysis of randomised controlled trials. BMC Cancer
Multimodality Imaging Evaluation of Adult Patients during and
2010;10: 337.
after Cancer Therapy: A Report from the American Society of
8. Moja L, Tagliabue L, Balduzzi S, et al. Trastuzumab containing Echocardiography and the European Association of Cardiovascular
regimens for early breast cancer. Cochrane Datab Syst Rev 2012; 4: Imaging. J Am Soc Echocardiogr 2014; 27: 911-939.
Cd006243.
19. Thavendiranathan P, Poulin F, Lim KD, et al. Use of myocardial
9. Malhotra V, Dorr VJ, Lyss AP, et al. Neoadjuvant and adjuvant strain imaging by echocardiography for the early detection of car-
chemotherapy with doxorubicin and docetaxel in locally advanced diotoxicity in patients during and after cancer chemotherapy: a
breast cancer. Clin Breast Cancer 2004;5 : 377-384. systematic review. J Am Coll Cardiol 2014; 63: 2751-2768.
10. Van Besien K, Devine S, Wickrema A, et al. Regimen related 20. Takigiku K, Takeuchi M, Izumi C, et al. Normal range of left
toxicity after fludarabine-melphalan conditioning: a prospective ventricular 2-dimensional strain: Japanese Ultrasound Speckle
study of 31 patients with hematologic malignancies. Bone Marrow Tracking of the Left Ventricle (JUSTICE) study. Circ J. 2012; 76:
Transplant 2003;32 : 471-476. 2623-2632.
11. Bovelli D, Platanitotis D, Roila F. ESMO Guidelines Working 21. Lenihan DJ, Massey MR, Baysinger KB, Adorno CL, Warneke CL,
Group. Cardiotoxicity of chemotherapeutic and radiotherapy- et al. Superior detection of cardiotoxicity during chemotherapy
related heart disease: ESMO Clinical Practice Guidelines. Ann using biomarkers. J Card Fail. 2007; 13 : S151.
Oncol 2010;21(Suppl5): v277-v282.
22. Ky B, Putt M, Sawaya H, et al. Early increases in multiple bio-
12. Heck SL, Gulati G, Ree AH, et al. Rationale and design of the markers predict subsequent cardiotoxicity in patients with breast
prevention of cardiac dysfunction during an adjuvant breast cancer cancer treted with doxorubicin, taxanes, and trastuzumab. J Am
therapy (PRADA) trial. Cardiology 2012; 123: 240-247. Coll Cardiol 2014; 63: 809-816.

Amit Varma, M.D., PhD, FACC


The Heart Group of Lancaster General Health
217 Harrisburg Ave.
Lancaster, PA 17603
717.544.8312
avarma2@lghealth.org

The Journal of Lancaster General Hospital • Winter 2017 • Vol. 12 – No. 4 111

You might also like