ArticlePDF Available

Monitoring the heart during cancer therapy

Authors:

Abstract and Figures

A growing number of effective cancer therapies is associated with cardiovascular (CV) toxicities including myocardial injury or dysfunction, leading to reduced ventricular function, and increased risk of heart failure. As the timing of administration of cancer treatment is known, the potential for risk stratification pre-treatment, and appropriate surveillance and monitoring during treatment, and intervention with cardio-protective treatment strategies in patients exhibiting early evidence of CV toxicity is an appealing clinical strategy. The field of cardio-oncology has developed, and the application of monitoring strategies using CV biomarkers and CV imaging has been to focus of many studies and is now implemented in dedicated cardio-oncology services supporting oncology centres. In this article, we review the background and rationale for monitoring, the different options and their strengths, weaknesses and where they are helpful in specific cardiotoxic cancer therapies, and the impact in cardio-oncology care.
Content may be subject to copyright.
Monitoring the heart during cancer therapy
Mohsen Habibian
1,2
and Alexander R. Lyon
2,3
*
1
The Prince Charles Hospital and University of Queensland, Rode Road, Chermside, QLD 4032, Australia;
2
Cardio-Oncology Service, Royal Brompton Hospital, Sydney Street, Chelsea, London, SW3 6NP, UK; and
3
National Heart and Lung Institute, Imperial College London, Cale Street, Chelsea, London, SW3 6LY, UK
KEYWORDS
Heart failure;
Cancer;
Cardio-oncology;
Monitoring
A growing number of effective cancer therapies is associated with cardiovascular (CV)
toxicities including myocardial injury or dysfunction, leading to reduced ventricular
function, and increased risk of heart failure. As the timing of administration of cancer
treatment is known, the potential for risk stratification pre-treatment, and appropriate
surveillance and monitoring during treatment, and intervention with cardio-protective
treatment strategies in patients exhibiting early evidence of CV toxicity is an appealing
clinical strategy. The field of cardio-oncology has developed, and the application of
monitoring strategies using CV biomarkers and CV imaging has been to focus of many
studies and is now implemented in dedicated cardio-oncology services supporting on-
cology centres. In this article, we review the background and rationale for monitoring,
the different options and their strengths, weaknesses and where they are helpful in
specific cardiotoxic cancer therapies, and the impact in cardio-oncology care.
Introduction
In recent years, cancer treatment is becoming more effec-
tive with the advent of novel agents targeting new path-
ways, receptors and activating the immune system to
target the cancer cells. These advances have improved the
rates of successful treatment and survival from cancer.
However, the rate of complications related to these thera-
pies has also increased.
1
Cardiovascular (CV) complications
are a common and important side effect which not only has
the potential to affect the quality of life and longevity in
cancer survivors but also leads to interruption of effective
anticancer treatments leading to worse cancer outcomes.
2
Cardiovascular diseases are frequently the leading cause of
non-cancer-related mortality. For example, survivors of
cancers diagnosed in childhood,
36
patients with breast
cancer
7
and many other cancers, the risk of developing CV
complications persists for years after treatment with
anthracycline chemotherapy (AC) or radiotherapy to the
chest.
8
During cancer therapy, patients who have pre-
existing CV disease and even CV risk factors, display a
higher risk of cardiotoxicity from AC and other cardiotoxic-
targeted cancer therapies [e.g. vascular endothelial
growth factor inhibitors (VEGFi), BCr-ABl inhibitors, pro-
teasome inhibitors (PIs), Raf-MEK inhibitors, and
Gonadotrophin-releasing hormone (GnRH) receptor ago-
nists]. Cancer treatment pathways are scheduled which
allows a unique opportunity for a baseline pre-treatment
risk assessment and monitoring of CV toxicity allowing
pre-emptive treatment.
9
This approach requires an under-
standing of the short- and long-term CV complications
encountered with each specific oncology treatment, paired
with appropriate cardiology knowledge, allowing for path-
way design, diagnosis, and management of cardiac compli-
cations resulting as a consequences of cancer treatment on
the CV system, or as a result of the direct involvement of
the cancer in the heart, in the new subspecialty of cardio-
oncology.
10
Establishing and developing such a specialized
field requires close collaboration between cardio-
oncologists, oncologists, and haemato-oncologists. This
multidisciplinary approach has been shown to yield better
prognostic outcomes in patients suffering from numerous
different types of cancer.
11,12
The purpose of this subspeci-
alty is to prevent the development and minimize the pro-
gression of CV complications by guiding treatment
therapies and interventions tailored to the patient.
*Corresponding author. Tel: þ44 (0) 207 352 8121 (ext 82396),
Fax: þ44 (0) 207 351 8776, Email: a.lyon@ic.ac.uk
Published on behalf of the European Society of Cardiology. V
CThe Author(s) 2019.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs licence
(http://creativecommons.org/licenses/by-nc-nd/4.0/), which permits non-commercial reproduction and distribution of the work, in
any medium, provided the original work is not altered or transformed in any way, and that the work is properly cited. For commercial
re-use, please contact journals.permissions@oup.com
European Heart Journal Supplements (2019) 21 (Supplement M), M44–M49
The Heart of the Matter
doi:10.1093/eurheartj/suz230
Downloaded from https://academic.oup.com/eurheartjsupp/article-abstract/21/Supplement_M/M44/5691328 by guest on 31 December 2019
Cancer drug therapies can affect many structures in the
heart, including myocardium [causing inflammatory or
non-inflammatory left ventricular dysfunction and heart
failure (HF)], conduction system (heart block), electro-
physiology (tachy- or bradyarrhythmias and QT-prolonga-
tion), pericardium, coronary arteries (raising issues like
myocardial infarction, vasospasm, or accelerated athero-
sclerosis), valves (especially after high-dose radiotherapy
to the valve tissue), the pulmonary circulation causing pul-
monary hypertension, and the systemic arterial and venous
circulation leading to arterial hypertension, peripheral vas-
cular disease, and venous thromboembolism. Chest-di-
rected radiotherapy can increase coronary artery disease
or cause fibrotic changes predominantly to the valves;
pericardium or myocardium and, therefore, monitoring in
survivors who have received high-dose radiation to the
heart should consider these multiple CV diseases. Cancer
patients should also have serial assessment of modifiable
CV risk factors (cholesterol, hypertension, diabetes, smok-
ing status, and body mass index) and the risks addressed
according to the CV prevention guidelines for high-risk
patients.
13
Monitoring for CV disease during and after cancer ther-
apy will be discussed in two sections (See Figure 1). In the
first part, the general considerations are provided for all
cancer patients. In the second section, cancer treatments
with specific CV side effect profiles, requiring targeted CV
screening, and follow-up with individualized surveillance
programme are discussed. The complexity of this field for
each cancer therapy with a CV toxicity profile and how
pathways of care for low-, moderate-, and high-risk
patients is beyond the scope of this article, and we refer
the reader to the forthcoming series of position statements
from the Heart Failure Association (HFA) of the European
Society of Cardiology which will present Baseline risk
stratification proformas, detailed biomarker and imaging
surveillance pathways and treatment strategies for a range
of effective cancer therapies with potential cardiotoxicity.
Baseline cardiovascular risk assessment and
cardio-oncology follow-up
A baseline risk assessment is advised for patients scheduled
to receive a cancer therapy with potential CV toxicity. This
should be provided by the oncology and haemato-oncology
services in co-ordination with their local cardiology or
cardio-oncology service.
Assessment of baseline CV risk before cancer therapies
which are potentially cardiotoxic is advisable. Baseline risk
is based on several patient-related factors, treatment-re-
lated factors, and cancer-related factors. Young children
or individuals older than 65years, pre-existing conven-
tional CV risk factors (hypertension, diabetes, dyslipidae-
mia, and smoker), and antecedent heart disease (like left
ventricular dysfunction or coronary artery disease)
increases the risk of CV complications. Additionally, type of
treatment used, cumulative dose (AC and trastuzumab),
combination therapy (such as AC and HER2 receptors or
dual immunotherapy agents use), or intensive or acceler-
ated regimens put the patientat higher risk of cardiac com-
plications. The type and location of the cancer are
important. For instance, mediastinal tumours requiring ra-
diotherapy increases the cardiac side effects if the heart is
in the radiation field.
14
Pathways should ensure high-risk patients are referred
and reviewed promptly by the cardiology or cardio-
oncology service and guidance on monitoring during the
planned cancer therapy is provided with a schedule person-
alized to the patient, their CV risk factors and pre-existing
CV disease, the proposed cancer treatment and the context
(metastatic vs. curative intent, prognosis with and without
cancer treatment, alternative non-cardiotoxic cancer ther-
apies). Cardiac symptom review before and during cancer
treatment are important in addition to monitoring cardiac
function and injury directly via biomarkersand imaging.
12
Imaging assessment
Echocardiography
Echocardiography is a first-line cardiac imaging technique
with advantages including its availability, assessment of
multiple elements of cardiac function beyond left ventricu-
lar ejection fraction (LVEF), relatively cheap cost, and
it is radiation-free allowing multiple scans safely.
Echocardiographic assessment of cardiotoxicity allows de-
tection during and in the 12 months after treatment, and in
high-risk patients may be used for intermittent surveillance
for life. Most focus has been on monitoring LVEF is important
in monitoring the cancer patients during or after the treat-
ment, and a pre-treatment echocardiography in the high-
risk patients provide a baseline for further comparison.
12
Cancer therapeutics-related cardiac dysfunction
(CTRCD) has a range of definitions, from oncology trials to
cardiology studies and registries, which has added confu-
sion to the field. The joint EACVI-ACC imaging position
Monitoring
ECG
Standard Echo
Advanced Echo: Strain
studies (GLS), Tissue
Doppler, 3D volumes
Stress Echo
CMR
Biomarkers
Baseline
Cardiovascular
Assessment
Early detecon of
cardiotoxicity
Diagnosis of
cancer or
evaluaon
of cancer
progress
Early signs of CTRCD:
Commence cardioprotecve
medicaons and connue
cancer treatment
Moderate CTRCD:
Opmisaon of cardiac
medicaon, provisional
interrupon of cancer
treatment and reassess
Severe CTRCD:
Interrupon of
cancer treatment,
cardiac
opmisaon,
reassess and
balance cancer
treatment,
prognosis and
cardiac risks
Figure 1 Summary of monitoring the heart receiving cancer treatment.
Monitoring the heart during cancer therapy M45
Downloaded from https://academic.oup.com/eurheartjsupp/article-abstract/21/Supplement_M/M44/5691328 by guest on 31 December 2019
statement proposed one definition, the LVEF is reduced by
more than 10% from the baseline or it decreased to the
value <53%, and this should be confirmed in two consecu-
tive echocardiography assessments with 2–3 weeks apart.
15
However, there are several limitations of this type of defi-
nition. Left ventricular (LV) volumes and LVEF are ideally
measured by Simpson’s biplane method. However, LVEF is
both load- and heart rate-dependent in many patients.
Intravascular volume in the cancer patients is quite vari-
able, as they sometimes receive large amount of intrave-
nous fluidsat the time of chemotherapy, or are dehydrated
due to vomiting or diarrhoea. Sympathetic tone varies and
may be elevated due to stress, anxiety, and/or pain. These
changes all affect LVEF and introduce variability and poten-
tial error. Cancer patients may have suboptimal acoustic
windows form prior chest surgery or radiotherapy.
Contrast-enhanced echocardiography is useful in more ac-
curate assessment of LVEF; however, the LV volume whilst
using contrast may be overestimated and contrast obscure
other detailed measurements. Three-dimensional ejection
fraction may help to improve accuracy of measurements
but cannot always be acquired due to image quality. When
three-dimensional echocardiography is performed, it is
critical to recognize that the normal references for the car-
diac chambers and LVEF is different from two-dimensional
measurements. Three-dimensional imaging is highly based
on the two-dimensional image quality and it is not helpful
in the patients with difficult acoustic windows.
Another technically challenging issue is that LVEF assess-
ment cannot identify early cardiotoxicity in treatments
like anthracyclines
16
and when depressed LVEF is diag-
nosed, significant myocardial injury has already occurred is
more likely to be irreversible.
17
Tissue Doppler assessment and diastolic function, includ-
ing septal and lateral E’, S’, and mitral annulus plane
systolic excursion M-mode (MAPSE) as well as mitral inflow
E waves measurements have all been studied in different
trials and can add incremental value in monitoring the
heart in cancer patients, and at times it has helped to iden-
tify the cardiotoxicity in earlier stages. Although there is
no definite cut-off point to determine normality for these
measurements in cancer patients, serial assessment may
identify early cardiotoxicity. The transmitral E/Aratio may
be an independent predictor of cardiotoxicity in some stud-
ies although this has not been reproduced in other
studies.
18
Speckle tracking, especially to measure left ventricular
global longitudinal strain (GLS) may be helpful in detecting
subclinical LV dysfunction, and the values should be com-
pared against the baseline and more studies are in process
to elucidate the details. Impairment in GLS <8% probably
does not have clinical consequences. However, worsening
of >15% has been proposed as a clinically significant, but in
the opinion of the authors, this is only relevant if it also
falls into the abnormal GLS range. In addition, it is critical
that serial monitoring is performed with echocardiography
machines from the same vendor so the scale remains the
same.
15
Emerging data suggest that GLS reduction may be
relevant in specific cancer populations including those re-
ceiving immune checkpoint inhibitors (ICIs) and paediatric
cancer survivors during long-term follow-up.
19,20
In some
cancer populations, a reduction in GLS appears to predict
future fall in LVEF, but as a more sensitive imaging tech-
nique it must be applied cautiously (e.g. effective oncology
treatments must not be stopped based on a reduction of LV
GLS alone). More studies are required to prove impairment
in GLS can change outcomes and this will be addressed in
one cancer population in the SUCCOUR trial.
Monitoring valvular disease in cancer survivors who have
been exposed to high doses of cardiac radiation may be ap-
propriate. Prospective studies are absent, partly due tothe
long latency between treatment exposure and clinically
significant valve disease. Five yearly echocardiographic
surveillance from 5 years post-treatment may be consid-
ered, with more frequent assessment if abnormalities of
valvular function are detected. During and after treat-
ment, if infective endocarditis is suspected, transoesopha-
geal echocardiography may be appropriate. Mitral
regurgitation may be primary due to radiation-related
damage, or secondary to LV dysfunction. A similar principle
is also relevant for tricuspid regurgitation as primary
or secondary to right ventricular (RV) dilatation.
Radiotherapy in younger patients causes direct valvular in-
jury with fibrotic changes in intracardiac valves causing
stenosis or regurgitation and monitoring valvular disease is
important in these patients. In older patients with pre-
existing valve disease, lower radiation doses may acceler-
ate the underlying disease. When valvular dysfunction is
diagnosed, more frequent monitoring with serial echocar-
diography is recommended.
Pericardial diseases are also more common in cancer
patients after radiation therapy to the heart, and echocar-
diography provides opportunity to assess pericardial effu-
sion or consequences like tamponade or constrictive
physiology with preload challenge if equivocal results at
rest are detected.
Stress echocardiography may be applied for several indi-
cations. First is in the assessment for flow-limiting coronary
disease in patients at moderate or high risk before major
cancer surgery or cancer treatments which may potentially
cause myocardial ischaemia, including fluorpyrimidines
(5-fluorouracil and capecitabine) or VEGFi.
21
Serial assess-
ment of contractile reserve has been studied but is not
validated for routine clinical practice although it may be
helpful in early detection of subclinical cardiac dysfunc-
tion. Stress echocardiography is not only valuable in CV
prognosis but provocatively it may also helpful in estima-
tion of non-cardiac cancer death.
22
Endomyocardial biopsy
In the past considered as the most specific modality for di-
agnosis of cancer therapeutics-related cardiotoxicity.
However, owing to its invasive nature and inherent risks, it
is considered generally as a last line of investigation, espe-
cially with the advent of other modalities for monitoring
the heart like advanced echocardiography and biomarkers.
However, it is becoming increasingly important for the di-
agnosis or exclusion of ICI-related myocarditis in borderline
cases with discordant biomarker and imaging findings to
guide future ICI treatment.
M46 M. Habibian and A.R. Lyon
Downloaded from https://academic.oup.com/eurheartjsupp/article-abstract/21/Supplement_M/M44/5691328 by guest on 31 December 2019
Cardiac magnetic resonance
Cardiac magnetic resonance (CMR) provides detailed as-
sessment of LV and RV function and is the best imaging mo-
dality to evaluate presence or absence of fibrosis,
infarction, myocarditis, and amyloidosis, as well as pericar-
dial anatomy and inflammation, and assessment of intra-
cardiac masses. However, CMR has several important
limitations, including cost, accessibility, acquisition time,
contraindications in the patients with implanted metal
works, quality in patients with fast heart rates, AF or fre-
quent ectopy, and difficulties in the patients suffering from
claustrophobia or advanced anxiety.
MUGA
Multiple-gated acquisition scan (MUGA) was initially the
standard modality for early assessment of serial LVEF and
detection of subclinical left ventricular dysfunction prior
to the clinical HF. It has the advantage of being reproduc-
ible and useful in the patients with difficult acoustic win-
dows which makes the transthoracic echocardiography
difficult and is widely available with costs comparable with
other modalities. However, MUGA has drawbacks, particu-
larly, the cumulative dose of radiation, and inability to pro-
vide information about other cardiac elements like LV
diastolic function, right ventricle, PA pressure, valvular
function, or left atrium dimensions.
23
Cardiac biomarkers
Cardiac biomarkers are readily and widely available with
high precision, accuracy, and clear cut-off points for a
range of cardiac diseases, e.g. acute coronary syndromes.
Cardiac biomarkers including cardiac troponin (cTn) and
natriuretic peptides [brain natriuretic peptide (BNP) and
N-terminal portion of proBNP (NT-proBNP)] have been stud-
ied for early detection of cardiac dysfunction.
24
Cardiac
troponin’s are markers of cardiac injury, and in many stud-
ies showing the patients receiving AC with elevated tropo-
nin levels are at higher risk of cardiac events and
developing cardiotoxicity, and the magnitude and duration
of this elevation are important in cardiac prognosis after
completing AC.
25
The value of cTn in newer cancer drugs
like trastuzumab in treatment of breast cancer has also
been explored, although many of these patients have also
received AC.
26
Brain natriuretic peptide is more sensitive
than cTn during trastuzumab, VEGFi, and PI at detecting
early LV dysfunction and predicting function clinical
cardiac adverse events. Emerging data suggest BNP or
NT-proBNP may also be helpful in detecting ICI-medicated
cardiotoxicity. In an unselected population of all CV toxic-
ity from ICI, only 46% of the patients had positive troponin
levels but all (100%) had elevated BNP. However, this may
reflect different clinical practice regarding biomarker
measurement and more detailed studies are required. In
high-risk patients scheduled to receive potentially cardio-
toxic cancer treatments known to cause HF regular moni-
toring of cardiac biomarkers including cTn and natriuretic
peptides (NPs) should be considered for early detection of
cardiac dysfunction.
Anthracycline chemotherapy
Anthracycline chemotherapy, including doxorubicin, epiru-
bicin, daunorubicin, idarubicin, and anthracenediones
(mitoxantrone and pixantrone) cause cumulative dose-
dependent cardiotoxicity and dysfunction, AC’s cause cardi-
omyocyte damage via increased reactive oxygen species
and topoisomerase-IIbinhibition.
27
At the cellular level, it
causes vacuolar deformation before myofibrillar dysfunction
and cellular apoptosis.
28
If left late this leads to irreversible
damage and hence early measurement of cTn is helpful to
detect early myocardial injury before irreversible left ven-
tricular systolic dysfunction (LVSD) and HF develops.
29,30
HER2-targeted therapy
Trastuzumab, pertuzumab, T-DM1, lapatinib, and neratinib
are the HER2-targeted therapies with inhibit the HER2 re-
ceptor and improve prognosis in HER2þearly invasive and/
or metastatic HERþbreast cancer. Heart disease initiated
cardiac HER2 expression in a mechanism for cardioprotec-
tion and stabilization, and hence HER2 inhibitors cause LV
and/or RV dysfunction. In most cases, HER2-targeted ther-
apy-mediated cardiac dysfunction is reversible by interrup-
tion of the therapy and administration of cardioprotective
medications. However, interruption of HER2 targeted ther-
apies may lead to worse cancer outcomes, particularly in
metastatic HER2þbreast cancer and so preventing inter-
ruptions through monitoring is an appealing strategy.
Regular monitoring with echocardiography for early detec-
tion is recommended and recent studies suggest two new
approaches. The first is to use baseline cTn pre-treatment
and serial surveillance of both GLS and NPs during treat-
ment as a sensitive strategy. The second is to continue tras-
tuzumab in patients where the LVEF falls to 40–49%
providing the patient is clinically stable and with imple-
mentation of appropriate cardioprotective medication.
31
Vascular endothelial growth factor inhibitors
Vascular endothelial growth factor inhibitors (VEGFi) have
a relatively higher rate of CV side effects, particularly, the
more non-specific tyrosine kinase inhibitors (e.g. Sunitinib
and Sorafenib). The most common complications include
arterial hypertension, ischaemic events, arrhythmias due
to QT-prolongation, and cardiac dysfunction, which may be
multifactorial, including both hypertensive HF and direct
cardiotoxicity, particularly, in patients with pre-existing LV
dysfunction or hypertrophy. Therefore, a baseline risk as-
sessment in all cancer patients scheduled to receive a
VEGFi is recommended, with regular cardiac monitoring in
moderate and high-risk patients using echocardiography
and NPs. Regular blood pressure monitoring with home
blood pressure diaries, and electrocardiograms (ECG’s) in
clinic to measure QTc intervals,are recommended.
BCr-ABl TKI’s
BCr-Abl TKIs are predominantly used in the treatment of
chronic myeloid leukaemia with Imatinib being the
Monitoring the heart during cancer therapy M47
Downloaded from https://academic.oup.com/eurheartjsupp/article-abstract/21/Supplement_M/M44/5691328 by guest on 31 December 2019
characteristic agent in this group, which has minimal car-
diac side effects. However, second-generation therapies
such as Dasatinib and Nilotinib, and the third-generation
BCr-Abl TKI Ponatinib have significant CV toxicities.
Nilotinib accelerates the atherosclerotic process and
causes accentuation in baseline CV risk factors, to the ex-
tent that peripheral arterial disease leading to limb ampu-
tation has been reported. It also causes QTc-interval
prolongation. Monitoring requires regular assessment of
standard CV risk factors, including cholesterol, HbA1c,
blood pressure, and ECGs to measure QTc. Dasatinib may
cause pulmonary hypertension and/or HF and serial echo-
cardiography to address both LV function and PA pressure is
recommended in moderate- and high-risk cases (baseline
PA elevation and baseline LV dysfunction). Dasatinib cause
pleural effusion in up to 28% of cases with an unknown
mechanism but it does not appear to be cardiac. Bosutinib
has lower rates of CV complications, all of which in the
studies could be managed medically without the need for
discontinuation of Bosutinib. Ponatinib, which also being
used in the trials for acute lymphoblastic leukaemia, has a
relatively high risk of CV toxicity. This includes an increased
risk of occlusive arterial and venous occlusive events.
Ponatinib also causes HF in a dose-dependent effect and
myocardial infarction. Cardiac and CV risk factor monitor-
ing should be performed to address these potential side
effects and optimize CV risk factor control.
32
RAF/MEK TKI’s
RAF and MEK inhibitors are pro-survival pathways in various
cancers including melanoma and thyroid cancers. In combi-
nation, they have improved survival in Raf-mutant mela-
noma and other Raf-mutant cancers. Recent data have
clarified that HF or LVSD is caused by combination Raf- and
MEK inhibitors in 5–10% of patients.
33
Regular monitoring
for early detection of cardiac dysfunction with serial echo-
cardiography, biomarkers, and ECG’s may be considered in
the patients receiving these agents but more research is
required.
Immune checkpoint inhibitors
Immune checkpoint inhibitors ICIs are increasingly used to
treat a variety of neoplasms with important benefits.
Immune checkpoint inhibitor-mediated cardiotoxicity oc-
curring most commonly in the first few cycles, with inflam-
matory CV toxicities including myocarditis, pericarditis,
and vasculitis. LV dysfunction including cardiogenic shock,
and malignant ventricular arrhythmias may occur in
patients with myocarditis with high mortality rates of 25–
50%.
3436
The optimal monitoring strategy has not been
identified, but ECG and biomarker (cTn and NP) monitoring
in the first few cycles may be helpful. Cardiac complica-
tions are more prevalent in combination immunotherapy
settings, for example, the combination of Nivolumab and
Ipilimumab, or an ICI plus a VEGFi. The effects of ICI’s are
variable in different individuals. They have the potential to
cause heart block, tachyarrhythmias, cardiac dysfunction,
pericarditis, acute coronary syndrome, or vasculitis.
Monitoring may consist of regular ECG’s, cardiac bio-
markers, and echocardiography. In case of LV dysfunction,
CMR is very important in diagnosing the underlying aetiol-
ogy and distinguishing the inflammatory from non-
inflammatory dysfunction, which has different treatment
options and implications in cancer therapy.
Ibrutinib
Ibrutinib is a first-generation Bruton Tyrosine Kinase inhibi-
tor used in B-cell cancers, demonstrating cardiotoxicity in-
cluding a high risk of new atrial fibrillation and a small
increased risk of ventricular arrhythmias and sudden car-
diac death.
37
Monitoring for new AF with serial ECGs, or
other technologies for cardiac rhythm monitoring, may be
considered.
Conclusion
Cardiovascular monitoring of selected cancer patients re-
ceiving cardiotoxic cancer treatments, who are at higher
risk, is a logical clinical strategy for the early detection of
cardiac dysfunction if this leads to implementation of car-
dioprotective strategies to allow effective cancer thera-
pies to continue safely. More trials are required to identify
the optimal monitoring strategy for each cancer treat-
ment, including which modalities (imaging, biomarkers,
and combination) and frequency, with the goal of improv-
ing both cancer and CV outcomes. Expert consensus and po-
sition papers from the HFA will address these challenges
and provide clinical pathways in the near future based on
the experience and limitedevidence available.
Conflict of interest: Dr A.R.L. received speaker, advisory board or
consultancy fees and/or research grants from Pfizer, Novartis,
Servier, Amgen, Takeda, Roche, Janssens-Cilag Ltd, Clinigen
Group, Eli Lily, Eisai, Bristol Myers Squibb, Ferring Pharmaceuticals
and Boehringer Ingelheim. M.H. has no conflicts to declare.
References
1. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh
JWW, Comber H, Forman D, Bray F. Cancer incidence and mortality
patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer
2013;49:1374–1403.
2. Zamorano JL, Lancellotti P, Rodriguez Munoz D, Aboyans V,
Asteggiano R, Galderisi M, Habib G, Lenihan DJ, Lip GYH, Lyon AR,
Lopez-Fernandez T, Mohty D, Piepoli MF, Tamargo J, Torbicki A, Suter
TM; ESC Scientific Document Group. 2016 ESC Position Paper on can-
cer treatments and cardiovascular toxicity developed under the aus-
pices of the ESC Committee for Practice Guidelines: the Task Force
for cancer treatments and cardiovascular toxicity of the European
Society of Cardiology (ESC). Eur J Heart Fail 2017;19:9–42.
3. Reulen RC, Winter DL, Frobisher C, Lancashire ER, Stiller CA, Jenney
ME, Skinner R, Stevens MC, Hawkins MM; British Childhood Cancer
Survivor Study Steering Group. Long-term cause-specific mortality
among survivors of childhood cancer. JAMA 2010;304:172–179.
4. Oeffinger KC, Mertens AC, Sklar CA, Kawashima T, Hudson MM,
Meadows AT, Friedman DL, Marina N, Hobbie W, Kadan-Lottick NS,
Schwartz CL, Leisenring W, Robison LL. Chronic health conditions in
adult survivors of childhood cancer. N Engl J Med 2006;355:1572–1582.
5. Mertens AC, Liu Q, Neglia JP, Wasilewski K, Leisenring W, Armstrong
GT, Robison LL, Yasui Y. Cause-specific late mortality among 5-year
survivors of childhood cancer: the Childhood Cancer Survivor Study.
J Natl Cancer Inst 2008;100:1368–1379.
M48 M. Habibian and A.R. Lyon
Downloaded from https://academic.oup.com/eurheartjsupp/article-abstract/21/Supplement_M/M44/5691328 by guest on 31 December 2019
6. Geenen MM, Cardous-Ubbink MC, Kremer LCM, van den Bos C, van
der Pal HJH, Heinen RC, Jaspers MWM, Koning CCE, Oldenburger F,
Langeveld NE, Hart AAM, Bakker PJM, Caron HN, van Leeuwen FE.
Medical assessment of adverse health outcomes in long-term survi-
vors of childhood cancer. JAMA 2007;297:2705–2715.
7. Patnaik JL, Byers T, DiGuiseppi C, Dabelea D, Denberg TD.
Cardiovascular disease competes with breast cancer as the leading
cause of death for older females diagnosed with breast cancer: a
retrospective cohort study. Breast Cancer Res 2011;13:64.
8. Fidler MM, Reulen RC, Henson K, Kelly J, Cutter D, Levitt GA,
Frobisher C, Winter DL, Hawkins MM. Population-based long-term
cardiac-specific mortality among 34 489 five-year survivors of child-
hood cancer in Great Britain. Circulation 2017;135:951–963.
9. Armstrong GT, Oeffinger KC, Chen Y, Kawashima T, Yasui Y,
Leisenring W, Stovall M, Chow EJ, Sklar CA, Mulrooney DA, Mertens
AC, Border W, Durand J-B, Robison LL, Meacham LR. Modifiable risk
factors and major cardiac events among adult survivors of childhood
cancer. J Clin Oncol 2013;31:3673–3680.
10. Hong RA, Iimura T, Sumida KN, Eager RM. Cardio-oncology/onco-car-
diology. Clin Cardiol 2010;33:733–737.
11. Lancellotti P, Suter TM, Lopez-Fernandez T, Galderisi M, Lyon AR,
Van der Meer P, Cohen Solal A, Zamorano JL, Jerusalem G, Moonen
M, Aboyans V, Bax JJ, Asteggiano R. Cardio-oncology services: ratio-
nale, organization, and implementation: a report from the ESC
Cardio-Oncology council. Eur Heart J 2019;40:1756–1763.
12. Pareek N, Cevallos J, Moliner P, Shah M, Tan LL, Chambers V, Baksi
AJ, Khattar RS, Sharma R, Rosen SD, Lyon AR. Activity and outcomes
of a cardio-oncology service in the United Kingdom-a five-year expe-
rience. Eur J Heart Fail 2018;20:1721–1731.
13. Piepoli MF, Hoes AW, Agewall S, Albus C, Brotons C, Catapano AL,
Cooney MT, Corra` U, Cosyns B, Deaton C, Graham I, Hall MS, Hobbs
FDR, Løchen ML, Lo¨llgen H, Marques-Vidal P, Perk J, Prescott E,
Redon J, Richter DJ, Sattar N, Smulders Y, Tiberi M, van der Worp
HB, van Dis I, Verschuren WMM, Binno S; ESC Scientific Document
Group. 2016 European Guidelines on cardiovascular disease preven-
tion in clinical practice: the Sixth Joint Task Force of the European
Society of Cardiology and Other Societies on Cardiovascular Disease
Prevention in Clinical Practice (constituted by representatives of 10
societies and by invited experts). Developed with the special contri-
bution of the European Association for Cardiovascular Prevention &
Rehabilitation (EACPR). Eur Heart J 2016;37:2315–2381.
14. Suter TM, Ewer MS. Cancer drugs and the heart: importance and
management. Eur Heart J 2013;34:1102–1111.
15. Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M,
Ganame J, Sebag IA, Agler DA, Badano LP, Banchs J, Cardinale D,
Carver J, Cerqueira M, DeCara JM, Edvardsen T, Flamm SD, Force T,
Griffin BP, Jerusalem G, Liu JE, Magalh~
aes A, Marwick T, Sanchez LY,
Sicari R, Villarraga HR, Lancellotti P. Expert consensus for multimo-
dality 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.
16. Ewer MS, Ali MK, Mackay B, Wallace S, Valdivieso M, Legha SS,
Benjamin RS, Haynie TP. A comparison of cardiac biopsy grades and
ejection fraction estimations in patients receiving Adriamycin. J Clin
Oncol 1984;2:112–117.
17. Jensen BV, Skovsgaard T, Nielsen SL. Functional monitoring of
anthracycline cardiotoxicity: a prospective, blinded, long-term ob-
servational study of outcome in 120 patients. Ann Oncol 2002;13:
699–709.
18. Bountioukos M, Doorduijn JK, Roelandt JR, Vourvouri EC, Bax JJ,
Schinkel AF, Kertai MD, Sonneveld P, Poldermans D. Repetitive dobut-
amine stress echocardiography for the prediction of anthracycline
cardiotoxicity. Eur J Echocardiogr 2003;4:300–305.
19. Armstrong GT, Joshi VM, Ness KK, Marwick TH, Zhang N, Srivastava
D, Griffin BP, Grimm RA, Thomas J, Phelan D, Collier P, Krull KR,
Mulrooney DA, Green DM, Hudson MM, Robison LL, Plana JC.
Comprehensive echocardiographic detection of treatment-related
cardiac dysfunction in adult survivors of childhood cancer: results
from the St. Jude Lifetime Cohort Study. J Am Coll Cardiol 2015;65:
2511–2522.
20. Portugal G, Moura Branco L, Galrinho A, Mota Carmo M, Timoteo AT,
Feliciano J, Abreu J, Duarte Oliveira S, Batarda L, Cruz Ferreira R.
Global and regional patterns of longitudinal strain in screening for
chemotherapy-induced cardiotoxicity. Rev Port Cardiol 2017;36:
9–15.
21. Yeh ET, Bickford CL. Cardiovascular complications of cancer therapy:
incidence, pathogenesis, diagnosis, and management. J Am Coll
Cardiol 2009;53:2231–2247.
22. Carpeggiani C, Landi P, Michelassi C, Andreassi MG, Sicari R, Picano
E. Stress echocardiography positivity predicts cancer death. JAm
Heart Assoc 2017;6:e007104.
23. Gottdiener JS, Mathisen DJ, Borer JS, Bonow RO, Myers CE, Barr LH,
Schwartz DE, Bacharach SL, Green MV, Rosenberg SA. Doxorubicin
cardiotoxicity: assessment of late left ventricular dysfunction by ra-
dionuclide cineangiography. Ann Intern Med 1981;94:430–435.
24. Cardinale D, Sandri MT. Role of biomarkers in chemotherapy-induced
cardiotoxicity. Prog Cardiovasc Dis 2010;53:121–129.
25. Cardinale D, Sandri MT, Colombo A, Colombo N, Boeri M, Lamantia
G, Civelli M, Peccatori F, Martinelli G, Fiorentini C, Cipolla CM.
Prognostic value of troponin I in cardiac risk stratification of cancer
patients undergoing high-dose chemotherapy. Circulation 2004;109:
2749–2754.
26. Cardinale D, Colombo A, Torrisi R, Sandri MT, Civelli M, Salvatici M,
Lamantia G, Colombo N, Cortinovis S, Dessanai MA, Nole` F, Veglia F,
Cipolla CM. Trastuzumab-induced cardiotoxicity: clinical and prog-
nostic implications of troponin I evaluation. J Clin Oncol 2010;28:
3910–3916.
27. Zhang S, Liu X, Bawa-Khalfe T, Lu L-S, Lyu YL, Liu LF, Yeh ETH.
Identification of the molecular basis of doxorubicin-induced cardio-
toxicity. Nat Med 2012;18:1639–1642.
28. Friedman MA, Bozdech MJ, Billingham ME, Rider AK. Doxorubicin car-
diotoxicity. Serial endomyocardial biopsies and systolic time inter-
vals. JAMA 1978;240:1603–1606.
29. Felker GM, Thompson RE, Hare JM, Hruban RH, Clemetson DE,
Howard DL, Baughman KL, Kasper EK. Underlying causes and long-
term survival in patients with initially unexplained cardiomyopathy.
N Engl J Med 2000;342:1077–1084.
30. Ewer MS, Lippman SM. Type II chemotherapy-related cardiac dys-
function: time to recognize a new entity. J Clin Oncol 2005;23:
2900–2902.
31. Lynce F, Barac A, Geng X, Dang C, Yu AF, Smith KL, Gallagher C,
Pohlmann PR, Nunes R, Herbolsheimer P, Warren R, Srichai MB,
Hofmeyer M, Cunningham A, Timothee P, Asch FM, Shajahan-Haq A,
Tan MT, Isaacs C, Swain SM. Prospective evaluation of the cardiac
safety of HER2-targeted therapies in patients with HER2-positive
breast cancer and compromised heart function: the SAFE-HEaRt
study. Breast Cancer Res Treat 2019;175:595–603.
32. Aghel N, Delgado DH, Lipton JH. Cardiovascular toxicities of BCR-
ABL tyrosine kinase inhibitors in chronic myeloid leukemia: preven-
tive strategies and cardiovascular surveillance. Vasc Health Risk
Manag 2017;13:293–303.
33. Mincu RI, Mahabadi AA, Michel L, Mrotzek SM, Schadendorf D, Rassaf
T, Totzeck M. Cardiovascular adverse events associated with BRAF
and MEK inhibitors: a systematic review and meta-analysis. JAMA
Netw Open 2019;2:e198890.
34. Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y,
Hicks M, Puzanov I, Alexander MR, Bloomer TL, Becker JR, Slosky DA,
Phillips EJ, Pilkinton MA, Craig-Owens L, Kola N, Plautz G, Reshef DS,
Deutsch JS, Deering RP, Olenchock BA, Lichtman AH, Roden DM,
Seidman CE, Koralnik IJ, Seidman JG, Hoffman RD, Taube JM, Diaz
LA, Anders RA, Sosman JA, Moslehi JJ. Fulminant myocarditis with
combination immune checkpoint blockade. N Engl J Med 2016;375:
1749–1755.
35. Moslehi JJ, Johnson DB, Sosman JA. Myocarditis with immune check-
point blockade. N Engl J Med 2017;376:292.
36. Lyon AR, Yousaf N, Battisti NML, Moslehi J, Larkin J. Immune check-
point inhibitors and cardiovascular toxicity. Lancet Oncol 2018;19:
e447–e458.
37. Woyach JA, Ruppert AS, Heerema NA, Zhao W, Booth AM, Ding W,
Bartlett NL, Brander DM, Barr PM, Rogers KA, Parikh SA, Coutre S,
Hurria A, Brown JR, Lozanski G, Blachly JS, Ozer HG, Major-Elechi B,
Fruth B, Nattam S, Larson RA, Erba H, Litzow M, Owen C, Kuzma C,
Abramson JS, Little RF, Smith SE, Stone RM, Mandrekar SJ, Byrd JC.
Ibrutinib regimens versus chemoimmunotherapy in older patients
with untreated CLL. N Engl J Med 2018;379:2517–2528.
Monitoring the heart during cancer therapy M49
Downloaded from https://academic.oup.com/eurheartjsupp/article-abstract/21/Supplement_M/M44/5691328 by guest on 31 December 2019
... stratification, and provide predictive insights for personalized cardiovascular care [20]. AI-driven decision support systems are being increasingly integrated into clinical workflows, helping clinicians detect cardiovascular risks earlier and tailor interventions more effectively [21][22][23]. Alongside AI, the development of smart cardiac devices has revolutionized real-time cardiovascular monitoring [24]. Wearable technologies, such as ECG patches, smartwatches, and biosensors, allow for the continuous, remote tracking of patients' cardiac health, reducing the need for in-hospital assessments [25]. ...
... In cardio-oncology, AI provides innovative solutions for the early detection of cardiotoxicity, risk stratification, and the optimization of therapeutic interventions [33]. The evolution of AI technologies has enabled the use of advanced ML algorithms and the processing of complex medical data, facilitating real-time data-driven clinical decision-making [22]. ...
... • Cardiotoxicity risk prediction: ML models can analyze clinical and imaging data to estimate the likelihood of cardiovascular complications before, during, or after oncology treatment [34]; • Real-time patient monitoring: Smart wearables and AI systems integrated into remote monitoring allow for the early detection of relevant physiological changes [23]; • Personalization of treatment: AI helps to adjust doses and choose the most appropriate therapeutic regimens for each patient, minimizing the risk of cardiac toxicity without compromising the efficacy of oncology treatment [22]; • Automated imaging analysis: DL algorithms can analyze echocardiograms, cardiac MRIs, and other medical images to identify structural or functional changes in the myocardium [35]; ...
Article
Full-text available
The increasing prevalence of cardiovascular complications in cancer patients due to cardiotoxic treatments has necessitated advanced monitoring and predictive solutions. Cardio-oncology is an evolving interdisciplinary field that addresses these challenges by integrating artificial intelligence (AI) and smart cardiac devices. This comprehensive review explores the integration of artificial intelligence (AI) and smart cardiac devices in cardio-oncology, highlighting their role in improving cardiovascular risk assessment and the early detection and real-time monitoring of cardiotoxicity. AI-driven techniques, including machine learning (ML) and deep learning (DL), enhance risk stratification, optimize treatment decisions, and support personalized care for oncology patients at cardiovascular risk. Wearable ECG patches, biosensors, and AI-integrated implantable devices enable continuous cardiac surveillance and predictive analytics. While these advancements offer significant potential, challenges such as data standardization, regulatory approvals, and equitable access must be addressed. Further research, clinical validation, and multidisciplinary collaboration are essential to fully integrate AI-driven solutions into cardio-oncology practices and improve patient outcomes.
... Our findings suggested that these markers could complement traditional biomarkers and can be combined with imaging techniques to create a more robust framework for early detection of AIC. 68,69 Finally, exercise interventions demonstrated significant potential in AIC prevention. Both aerobic exercise and resistance training showed beneficial effects on cardiovascular outcomes in patients undergoing anthracycline treatment. ...
... On the other hand, MAPSE measurement does not require good image quality (because it is a linear measurement that is less affected by artefacts) or multiple views and is not a time-consuming technique, thus making its measurement extremely useful in case of emergency settings and poor sonographic windows [6]. There has historically been ample data published on its usefulness in evaluating the longitudinal systolic function of the LV in heart failure (HF); however, recent publications have raised the question of its validity and use as a sensitive marker of early LV dysfunction and as a prognostic tool not only for HF patients, but also in the setting of septic shock, cardio-oncology, and many others [7,8]. There appears to be a lack of validation of its use in the context of conventional, endocardial right ventricular pacing ( Figure 2). ...
Article
Full-text available
Mitral annular plane systolic excursion (MAPSE) was a widely used and simple M-mode echocardiographic parameter for determining the left ventricle (LV) longitudinal systolic function. The purpose of this review is to analyze the use of MAPSE as a simple LV systolic function marker in different clinical scenarios, especially given the recent paradox of choices in ultrasound markers assessing cardiac performance. Recent data on the use of MAPSE in the assessment of LV function in different settings seem to be relatively scarce, given the wide variety of possible causes of cardiovascular pathology. There remain significant possible clinical applications of MAPSE utilization. This review included all major articles on the topic of mitral annular plane systolic excursion published and indexed in the PubMed, Google Scholar, and Scopus databases. We analyzed the potential implications of using simpler ultrasonographical tools in heart failure diagnosis, prediction, and treatment. MAPSE is a dependable, robust, and easy-to-use parameter compared to ejection fraction (EF) or global longitudinal strain (GLS) for the quick assessment of LV systolic function in various clinical settings. However, there may be a gap of evidence in certain scenarios such as conventional cardiac pacing.
... CTRCD is generally defined as a decrease in GLS of ≥ 15% from baseline or a decline in LVEF of ≥ 10% to an absolute LVEF of < 50%-53%, although the cut-off values vary according to guidelines [18,19]. If CTRCD is suspected, a follow-up TTE for confirmation is recommended within 2 to 3 weeks because the measurements are load-dependent [20,21]. Additionally, GLS values are vendor-dependent; thus, follow-up TTE should be performed with the same equipment. ...
Article
Full-text available
The global incidence of breast cancer is on the rise, a trend also observed in South Korea. However, thanks to the rapid advancements in anticancer therapies, survival rates are improving. Consequently, post-treatment health and quality of life for breast cancer survivors are emerging as significant concerns, particularly regarding treatment-related cardiotoxicity. In this review, we delve into the cardiovascular complications associated with breast cancer treatment, explore surveillance protocols for early detection and diagnosis of late complications, and discuss protective strategies against cardiotoxicity in breast cancer patients undergoing anticancer therapy, drawing from multiple guidelines.
... In cancer patients, there is a high incidence of cardiomyopathy-related mutations, such as the DNA damage response/repair system, and mutations in the DNA damage response/repair system gene increase the risk of cardiotoxicity with anticancer therapy [41]. Meanwhile, pathophysiological alterations in hereditary cardiomyopathy can promote cancer development and progression and may further increase the cardiotoxic effects of anticancer therapies [30,42,43]. ...
Article
Full-text available
Background: The purpose of this study was to evaluate the risk of cardiovascular mortality (CVM) among patients with bladder cancer (BC). Methods and Materials: Data were collected from the Surveillance, Epidemiology, and End Results (SEER) database for patients who were diagnosed with BC by pathology between 2000 and 2016. The standardized mortality rate (SMR) was calculated based on reference data from the general population. Nelson–Aalen cumulative hazard curves were used to assess the risk of experiencing CVM in BC patients. Multivariate competing risk models were performed. Results: In total, data from 237,563 BC patients were obtained from the SEER database for further analysis, of which 21,822 patients experienced CVM; the overall SMR for CVM in BC patients was 1.16 (95% CI: 1.14–1.17). Age, race, sex, year of diagnosis, histologic type, summary stage, surgery, marital status, and college education level were independent predictors of CVM in patients with BC. Conclusions: Patients with BC have a significantly increased risk of experiencing CVM compared to the general population. Pre-identification of high-risk groups and cardiovascular protection interventions are important measures to effectively improve survival in this group of patients.
... [2][3][4][5][6] Cancer therapy-related cardiac dysfunction (CTRCD) has become the second leading cause of cancer-related mortality. [7][8][9] The causes of CTRCD may be related to oxidative stress, endoplasmic reticulum stress, and mitochondrial dysfunction, but the mechanistic details are not known. [2][3][4][5]10 Recently, heart failure has been linked to increased expression of p53, epigenetic changes such as histone modifications, and DNA methylation, 11 but the relationship of these factors with CTRCD is not clear. ...
Article
Full-text available
Aims Cancer therapy‐related cardiac dysfunction (CTRCD) is commonly reported, but its histopathology, mechanisms, and risk factors are not known. We aimed to clarify the histopathology and mechanisms of CTRCD to identify risk factors. Methods and results We performed myocardial histopathological studies on 13 endomyocardial biopsies from CTRCD patients, 35 autopsied cancer cases with or without cardiac dysfunction, and controls without cancer (10 biopsies and 9 autopsies). Cardiotoxicity risk scores were calculated based on medication; and patient‐related risk factors, fibrosis, and cardiomyocyte changes were scored; and p53 and H3K27ac histone modification were evaluated by histological score (H‐score). In the biopsy cases, all histopathological changes and the p53 evaluation were significantly higher in the CTRCD group than in the controls [p53 H‐score; 63 (9.109) vs. 33 (5.099), P < 0.05]. In patients with a short time between drug and disease onset (<4.2 years), fibrosis and p53 positively correlated (r = 0.76, P < 0.05), and in those with late onset disease (>4.2 years), cellular abnormalities and p53 trended to a positive correlation and cardiotoxicity risk scores and p53 positively correlated (r = 0.95, P < 0.05). A year after biopsy, the short‐term group had significant recovery of ejection fraction compared with the long‐term group (P < 0.05). The CTRCD group had a significantly worse overall survival prognosis than the control group [hazard ratio 7.61 (95% confidence interval 1.30–44.6), P < 0.05]. Autopsy cases with cancer treatment also had a high grade of histopathological changes, with even more severe changes in patients with cardiac dysfunction, and had increased p53 and H3K27ac expression levels, compared with controls. H‐scores of p53 and H3K27ac showed a positive correlation in the CTRCD group in biopsy cases (r = 0.62, P < 0.05) and a positive correlation in autopsy cases. Conclusions Our results indicate distinct morphological characteristics in myocardial histopathology associated with CTRCD. p53 and H3K27ac histone modification could be sensitive markers of CTRCD and suggest a mechanistic involvement of epigenetic changes.
... However, in the case of inhibitors with lower or unclear risk (e.g., imatinib), monitoring is required only in patients with predisposing conditions or symptoms suggesting cardiac disease. Initiation of treatment in patients with LVEF <50% should be avoided, and discontinuation of treatment is necessary in case of LVEF reduction [10,[117][118][119]. ...
Article
Full-text available
Kinase inhibitors (KIs) represent a growing class of drugs directed at various protein kinases and used in the treatment of both solid tumors and hematologic malignancies. It is a heterogeneous group of compounds that are widely applied not only in different types of tumors but also in tumors that are positive for a specific predictive factor. This review summarizes common cardiotoxic effects of KIs, including hypertension, arrhythmias with bradycardia and QTc prolongation, and cardiomyopathy that can lead to heart failure, as well as less common effects such as fluid retention, ischemic heart disease, and elevated risk of thromboembolic events. The guidelines for cardiac monitoring and management of the most common cardiotoxic effects of protein KIs are discussed. Potential signaling pathways affected by KIs and likely contributing to cardiac damage are also described. Finally, the need for further research into the molecular mechanisms underlying the cardiovascular toxicity of these drugs is indicated.
Article
Full-text available
Respiratory diseases and cardiovascular diseases (CVDs) have high prevalence and share common risk factors. In some respiratory diseases such as sleep apnoea and COPD, the evidence of their negative impact on the prognosis of CVDs seems clear. However, in other diseases it is less evident whether there is any direct relationship. With this in mind, our objective was to provide information that may be helpful to better understand the relationship between respiratory pathology and CVDs. There are different reasons for this relationship, such as shared risk factors, common pathophysiological mechanisms, side effects of treatment and the direct effect in the heart and great vessels of respiratory diseases. Indeed, aging and smoking are risk factors for CVDs and also for respiratory diseases such as obstructive sleep apnea (OSA), COPD and interstitial lung diseases (ILD). Furthermore, there are common pathophysiological mechanisms that affect both respiratory diseases and CVDs, such as accelerated atherosclerosis, microvascular dysfunction, endothelial dysfunction, inflammation, hypoxemia and oxidative stress. Besides that, it is well known that lung cancer, sarcoidosis and amyloidosis may directly affect the heart and great vessels. Finally, side effects of drugs for respiratory diseases and the discontinuation of treatments that are necessary for CVDs, such as β-blockers and aspirin, may have a deleterious impact on the cardiovascular system. In conclusion, the coexistence of respiratory diseases and CVDs is very common. It makes modifying diagnostic and therapeutic management necessary and is also a relevant prognostic factor.
Article
Oral oncolytic treatment options have expanded over the last decade and have brought to light the need to monitor and manage cardiovascular (CV) disease in patients being treated with these therapies. There is a need to assess CV risk before patients receive oral oncolytic therapy with known potential to cause negative CV sequelae such as left ventricular dysfunction, hypercholesterolemia, hypertension, and arrhythmias. The review highlights the need to evaluate traditional CV risk factors and their association with the development and progression of cancer. Additionally, this review suggests approaches to monitor for CV adverse events and manage CV disease during and after treatment with oral oncolytic therapy. Key guideline recommendations are reviewed and highlight specific approaches to minimize CV harm for patients exposed to oral oncolytic therapy. Careful monitoring and patient-centered decision making is key in choosing appropriate therapies. A multidisciplinary approach between oncologists, cardio-oncologists, pharmacists, and other members of the healthcare team is essential in navigating cardiac toxicities.
Article
Chemotherapy-induced cardiac toxicity is an undesirable yet very common effect that increases the risk of death and reduce the quality of life of individuals undergoing chemotherapy. However, no feasible methods and techniques are available to monitor and detect the degree of cardiotoxicity at an early stage. Therefore, in this project, we aim to develop a fluorescent nanoprobe to image the toxicity within the cardiac tissue induced by an anticancer drug. We have observed that vascular cell adhesion molecule 1 (VCAM1) protein alone with collagen was overly expressed within the heart, when an animal was treated with doxorubicin (DOX), because of inflammation in the epithelial cells. We hypothesize that developing a VCAM1-targeted peptide-based (VHPKQHRGGSKGC) fluorescent nanoprobe can detect and visualize the affected heart. In this regard, we prepared a poly(lactic-co-glycolic acid) (PLGA) nanoparticle linked with VCAM1 peptide and rhodamine B (PLGA−VCAM1−RhB). Selective binding and higher accumulation of the PLGA−VCAM1−RhB nanoprobes were detected in DOX-treated human cardiomyocyte cells (HCMs) compared to the untreated cells. For in vivo studies, DOX (5 mg/kg) was injected via the tail vein once in two weeks for 6 weeks (3 injection total). PLGA−VCAM1−RhB and PLGA−RhB were injected via the tail vein after 1 week of the last dose of DOX, and images were taken 4 h after administration. A higher fluorescent signal of PLGA−RhB−VCAM-1 (48.62% ± 12.79%) was observed in DOX-treated animals compared to the untreated control PLGA−RhB (10.61% ± 4.90) within the heart, indicating the specificity and targeting ability of PLGA−VCAM1−RhB to the inflamed tissues. The quantified fluorescence intensity of the homogenized cardiac tissue of PLGA−RhB−VCAM1 showed 156% higher intensity than the healthy control group. We conclude that PLGA−VCAM1−RhB has the potential to bind inflamed cardiac cells, thereby detecting DOX-induced cardiotoxicity and damaged heart at an early stage.
Article
Full-text available
Importance Cardiovascular adverse events (CVAEs) after treatment with BRAF and MEK inhibitors in patients with melanoma remain incompletely characterized. Objective To determine the association of BRAF and MEK inhibitor treatment with CVAEs in patients with melanoma compared with BRAF inhibitor monotherapy. Data Sources PubMed, Cochrane, and Web of Science were systematically searched for keywords vemurafenib, dabrafenib, encorafenib, trametinib, binimetinib, and cobinimetinib from database inception through November 30, 2018. Study Selection Randomized clinical trials reporting on CVAEs in patients with melanoma being treated with BRAF and MEK inhibitors compared with patients with melanoma being treated with BRAF inhibitor monotherapy were selected. Data Extraction and Synthesis Data assessment followed the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. Pooled relative risks (RRs) and 95% CIs were determined using random-effects and fixed-effects analyses. Subgroup analyses were conducted to assess study-level characteristics associated with CVAEs. Main Outcomes and Measures The selected end points were pulmonary embolism, a decrease in left ventricular ejection fraction, arterial hypertension, myocardial infarction, atrial fibrillation, and QTc interval prolongation. All-grade and high-grade (≥3) CVAEs were recorded. Results Overall, 5 randomized clinical trials including 2317 patients with melanoma were selected. Treatment with BRAF and MEK inhibitors was associated with an increased risk of pulmonary embolism (RR, 4.36; 95% CI, 1.23-15.44; P = .02), a decrease in left ventricular ejection fraction (RR, 3.72; 95% CI, 1.74-7.94; P < .001), and arterial hypertension (RR, 1.49; 95% CI, 1.12-1.97; P = .005) compared with BRAF inhibitor monotherapy. The RRs for myocardial infarction, atrial fibrillation, and QTc prolongation were similar between the groups. These results were consistent when assessing high-grade CVAEs (left ventricular ejection fraction: RR, 2.79; 95% CI, 1.36-5.73; P = .005; I² = 29%; high-grade arterial hypertension: RR, 1.54; 95% CI, 1.14-2.08; P = .005; I² = 0%), but RRs for high-grade pulmonary embolism were similar between groups. A higher risk of a decrease in left ventricular ejection fraction was associated with patients with a mean age younger than 55 years (RR, 26.50; 95% CI, 3.58-196.10; P = .001), and the associated risk of pulmonary embolism was higher for patients with a mean follow-up time longer than 15 months (RR, 7.70; 95% CI, 1.40-42.12; P = .02). Conclusions and Relevance Therapy with BRAF and MEK inhibitors was associated with a higher risk of CVAEs compared with BRAF inhibitor monotherapy. The findings may help to balance between beneficial melanoma treatment and cardiovascular morbidity and mortality.
Article
Full-text available
Purpose HER2-targeted therapies have substantially improved the outcome of patients with breast cancer, however, they can be associated with cardiac toxicity. Guidelines recommend holding HER2-targeted therapies until resolution of cardiac dysfunction. SAFE-HEaRt is the first trial that prospectively tests whether these therapies can be safely administered without interruptions in patients with cardiac dysfunction. Methods Patients with stage I–IV HER2-positive breast cancer candidates for trastuzumab, pertuzumab or ado-trastuzumab emtansine (TDM-1), with left ventricular ejection fraction (LVEF) 40–49% and no symptoms of heart failure (HF) were enrolled. All patients underwent cardiology visits, serial echocardiograms and received beta blockers and ACE inhibitors unless contraindicated. The primary endpoint was completion of the planned HER2-targeted therapies without developing either a cardiac event (CE) defined as HF, myocardial infarction, arrhythmia or cardiac death or significant asymptomatic worsening of LVEF. The study was considered successful if planned oncology therapy completion rate was at least 30%. Results Of 31 enrolled patients, 30 were evaluable. Fifteen patients were treated with trastuzumab, 14 with trastuzumab and pertuzumab, and 2 with TDM-1. Mean LVEF was 45% at baseline and 46% at the end of treatment. Twenty-seven patients (90%) completed the planned HER2-targeted therapies. Two patients experienced a CE and 1 had an asymptomatic worsening of LVEF to ≤ 35%. Conclusion This study provides safety data of HER2-targeted therapies in patients with breast cancer and reduced LVEF while receiving cardioprotective medications and close cardiac monitoring. Our results demonstrate the importance of collaboration between cardiology and oncology providers to allow for delivery of optimal oncologic care to this unique population.
Article
Full-text available
Background: Stress echocardiography (SE) predicts cardiac death, but an increasing share of cardiac patients eventually die of cancer. The aim of the study was to assess whether SE positivity predicts cancer death. Methods and results: In a retrospective analysis of prospectively acquired single-center, observational data, we evaluated 4673 consecutive patients who underwent SE from 1983 to 2009. All patients were cancer-free at index SE and were followed up for a median of 131 months (interquartile range 134). We separately analyzed predetermined end points: cardiovascular, cancer, and noncardiovascular, noncancer death, with and without competing risk. SE was positive in 1757 and negative in 2916 patients; 869 cardiovascular, 418 cancer, and 625 noncardiovascular, noncancer deaths were registered. The 25-year mortality was higher in SE-positive than in SE-negative patients, considering cardiovascular (40% versus 31%; P<0.001) and cancer mortality (26% versus 17%; P<0.01). SE positivity was a strong predictor of cancer (cause-specific hazard ratio 1.19; 95% confidence interval, 1.16-1.73; P=0.05) and cardiovascular mortality (1.18; 95% confidence interval, 1.03-1.35; P=0.02). Fine-Gray analysis to account for competing risk gave similar results. Cancer risk diverged after 15 years, whereas differences were already significant at 5 years for cardiovascular risk. Conclusions: SE results predict cardiovascular and cancer mortality. SE may act as a proxy of the shared risk factor milieu for cancer or cardiovascular death.
Article
Full-text available
Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment and outcomes of chronic myeloid leukemia (CML). Despite their significant impact on the management of CML, there is growing evidence that TKIs may cause cardiovascular and/or metabolic complications. In this review, we present the current evidence regarding the cardiovascular safety profiles of BCR-ABL TKIs. Methodological challenges of studies that reported the cardiovascular safety of TKIs are discussed. We also propose management strategies for cardiovascular surveillance and risk factor modification during treatment with these agents.
Article
Background Ibrutinib has been approved by the Food and Drug Administration for the treatment of patients with untreated chronic lymphocytic leukemia (CLL) since 2016 but has not been compared with chemoimmunotherapy. We conducted a phase 3 trial to evaluate the efficacy of ibrutinib, either alone or in combination with rituximab, relative to chemoimmunotherapy. Methods Patients 65 years of age or older who had untreated CLL were randomly assigned to receive bendamustine plus rituximab, ibrutinib, or ibrutinib plus rituximab. The primary end point was progression-free survival. The Alliance Data and Safety Monitoring Board made the decision to release the data after the protocol-specified efficacy threshold had been met. Results A total of 183 patients were assigned to receive bendamustine plus rituximab, 182 to receive ibrutinib, and 182 to receive ibrutinib plus rituximab. Median progression-free survival was reached only with bendamustine plus rituximab. The estimated percentage of patients with progression-free survival at 2 years was 74% with bendamustine plus rituximab and was higher with ibrutinib alone (87%; hazard ratio for disease progression or death, 0.39; 95% confidence interval [CI], 0.26 to 0.58; P<0.001) and with ibrutinib plus rituximab (88%; hazard ratio, 0.38; 95% CI, 0.25 to 0.59; P<0.001). There was no significant difference between the ibrutinib-plus-rituximab group and the ibrutinib group with regard to progression-free survival (hazard ratio, 1.00; 95% CI, 0.62 to 1.62; P=0.49). With a median follow-up of 38 months, there was no significant difference among the three treatment groups with regard to overall survival. The rate of grade 3, 4, or 5 hematologic adverse events was higher with bendamustine plus rituximab (61%) than with ibrutinib or ibrutinib plus rituximab (41% and 39%, respectively), whereas the rate of grade 3, 4, or 5 nonhematologic adverse events was lower with bendamustine plus rituximab (63%) than with the ibrutinib-containing regimens (74% with each regimen). Conclusions Among older patients with untreated CLL, treatment with ibrutinib was superior to treatment with bendamustine plus rituximab with regard to progression-free survival. There was no significant difference between ibrutinib and ibrutinib plus rituximab with regard to progression-free survival. (Funded by the National Cancer Institute and Pharmacyclics; ClinicalTrials.gov number, NCT01886872.)
Article
Aims Cardio‐oncology clinics optimise the cardiovascular status of cancer patients but there is a limited description of their structure, case mix, activity and results. The purpose of this paper is to describe the activity and outcomes of a cardio‐oncology service, particularly with respect to supporting optimal cancer treatment and survival. Methods and results We prospectively studied patients referred to our service from February 2011 to February 2016. New York Heart Association (NYHA) class and parameters of cardiac function were measured at baseline and after optimisation by our service. Up‐titration of cardiac treatment, continuation of cancer therapy and mortality were used as outcome measures. Of the 535 patients (55.8% females) referred, rates of cardiotoxicity for anthracyclines, anti‐HER2 agents and tyrosine kinase inhibitors were 75.8%, 69.8% and 62.1%, respectively. Patients with left ventricular systolic dysfunction (LVSD) (n =128) were younger, had higher rates of hypertension and previous exposure to chemotherapy/radiotherapy (P < 0.001). At a median follow‐up of 360 days, 93.8% of the patients with LVSD showed improvement in left ventricular ejection fraction (45% pre vs. 53% post; P < 0.001) and NYHA class (NYHA III–IV in 22% pre vs. 10% post; P = 0.01). All patients with normal left ventricular ejection fraction and biochemical or functional myocardial toxicity and 88% of patients with LVSD were deemed fit for continuation of cancer therapy after cardiovascular optimisation. Conclusions Through the establishment of a cardio‐oncology service, it is feasible to achieve high rates of cardiac optimisation and cancer treatment continuation.
Article
Immune checkpoint inhibitors are a new class of anticancer therapies that amplify T-cell-mediated immune responses against cancer cells. Immune checkpoint inhibitors have shown important benefits in phase 3 trials, and several agents have been approved for specific malignancies. Although adverse events from immune checkpoint inhibitors are a common occurrence, cardiotoxic effects are uncommon, but are often serious complications with a relatively high mortality. Most cardiotoxic effects appear to be inflammatory in nature. Clinical assessment of a combination of biomarkers, electrocardiography, cardiac imaging, and endomyocardial biopsy can be used to confirm a possible diagnosis. In this Review, we discuss the epidemiology of immune checkpoint inhibitor-mediated cardiotoxic effects, as well as their clinical presentation, subtypes, risk factors, pathophysiology, and clinical management, including the introduction of a new surveillance strategy.
Article
Aims: Anticancer therapies have extended the lives of millions of patients with malignancies, but for some this benefit is tempered by adverse cardiovascular (CV) effects. Cardiotoxicity may occur early or late after treatment initiation or termination. The extent of this cardiotoxicity is variable, depending on the type of drug used, combination with other drugs, mediastinal radiotherapy, the presence of CV risk factors, and comorbidities. A recent position paper from the European Society of Cardiology addressed the management of CV monitoring and management of patients treated for cancer. Methods and results: The current document is focused on the basis of the Cardio-Oncology (C-O) Services, presenting their rationale, organization, and implementation. C-O Services address the spectrum of prevention, detection, monitoring, and treatment of cancer patients at risk of cardiotoxicity and/or with concomitant CV diseases. These services require a multidisciplinary approach, with the aims of promoting CV health and facilitating the most effective cancer therapy. Conclusion: The expected growing volume of patients with cancer at risk of developing/worsening CV disease, the advent of new technological opportunities to refine diagnosis, and the necessity of early recognition of cancer therapy-related toxicity mandate an integrative multidisciplinary approach and care in a specialized environment. This document from the ESC Cardio-Oncology council proposes the grounds for creating C-O Services in Europe based on expert opinion.