The incidence of acute myeloid leukemia (AML) increases with age. Intensive induction chemotherapy containing cytarabine and an anthracycline has been part of the upfront and salvage treatment of AML for decades. Anthracyclines are associated with a significant risk of cardiotoxicity (especially anthracycline-related left ventricular dysfunction [ARLVD]). In the older adult population, the higher prevalence of cardiac comorbidities and risk factors may further increase the risk of ARLVD. In this article of the Young International Society of Geriatric Oncology group, we review the prevalence of ARLVD in patients with AML and factors predisposing to ARLVD, focusing on older adults when possible. In addition, we review the assessment of cardiac function and management of ARLVD during and after treatment. It is worth noting that only a minority of clinical trials focus on alternative treatment strategies in patients with mildly declined left ventricular ejection fraction or at a high risk for ARLVD. The limited evidence for preventive strategies to ameliorate ARLVD and alternative strategies to anthracycline use in the setting of cardiac comorbidities are discussed. Based on extrapolation of findings from younger adults and nonrandomized trials, we recommend a comprehensive baseline evaluation of cardiac function by imaging, cardiac risk factors, and symptoms to risk stratify for ARLVD. Anthracyclines remain an appropriate choice for induction although careful risk-stratification based on cardiac disease, risk factors, and predicted chemotherapy-response are warranted. In case of declined left ventricular ejection fraction, alternative strategies should be considered.

More than 60% of acute myeloid leukemia (AML) cases are diagnosed in adults aged ≥60 years.1  Anthracyclines have been part of the upfront and salvage treatment of AML since the 1970s.2  In the upfront setting, anthracycline is traditionally given over 3 days (eg, daunorubicin 45-90 mg/m2 per day, idarubicin 12 mg/m2 per day) in combination with cytarabine (100-200 mg/m2 per day continuously over 7 days) (“7+3” regimen). Other strategies include the use of mitoxantrone and/or high-dose cytarabine (1-3 g/m2). Anthracyclines are associated with cardiotoxicity; they can decrease left ventricular ejection fraction (LVEF) and contribute to the subsequent development of heart failure (HF). Compared with younger patients, anthracycline use in older patients with AML may be more challenging because of a higher prevalence of preexisting left ventricular dysfunction and an overall decreased response rate toward chemotherapy. In this narrative review, we provide an overview on anthracycline-related cardiotoxicity in older patients with AML.

Anthracycline-induced cardiotoxicity is generally divided into acute vs chronic, the latter of which is more common and occurs in a dose-dependent manner.3,4  Acute cardiotoxicity is typically not dose-dependent and may present as acute HF, arrhythmia, or myocarditis.5-8  The spectrum of clinical presentation of chronic cardiotoxicity ranges from subclinical LVEF decline to HF. Cardiotoxicity is frequently found following anthracycline use and is generally defined as >10% decrease in LVEF to final LVEF <50%.3,9,10  However, the exact cutoff values for decline in LVEF vary in published studies.11,12  Other criteria such as decreased left fractional shortening, abnormal wall motion, global longitudinal strain, and diastolic dysfunction have also been occasionally used to define anthracycline-related cardiotoxicity but they are currently not integrated into the standard assessment and definition.9,13,14  For the purpose of this article, we focus on anthracycline-related left ventricular dysfunction (ARLVD) that presents as a decline in LVEF.

There are multiple processes that contribute to development of ARLVD (Figure 1).15  After cellular uptake, daunorubicin is intercalated into mitochondrial and nuclear DNA. This causes DNA double-strand breaks and activates topoisomerase-2β, which induces apoptosis and cellular death. It also causes mitochondrial dysfunction through the formation of reactive oxygen species and endoplasmic reticulum stress. These processes contribute to the loss of functional cardiomyocytes, myocardial disarray, and development of interstitial fibrosis.15-17 

Figure 1.

Mechanisms of anthracycline-related left ventricular dysfunction. The development of anthracycline-related left-ventricular dysfunction is multifactorial.15  After injection, daunorubicin is rapidly distributed to various tissues (eg, heart, lung, kidneys, spleen, liver, lean tissue). After cellular uptake, it is intercalated into mitochondrial and nuclear DNA. This causes double-strand breaks and activates topoisomerase-2β, which induces apoptosis and cellular death. It also causes mitochondrial dysfunction through the formation of reactive oxygen species (ROS) and endoplasmic reticulum (ER) stress. Other contributing mechanisms include titin proteolysis and inhibition of the neuregulin/ErbB pathway.15-17  All these processes contribute to the loss of functional cardiomyocytes, myocardial disarray, and development of interstitial fibrosis leading to left ventricular dysfunction and finally chronic heart failure.15,17,68  The histopathological pictures are a courtesy of Karin Klingel, Department of Cardiopathology, University Hospital Tübingen (Tübingen, Germany).

Figure 1.

Mechanisms of anthracycline-related left ventricular dysfunction. The development of anthracycline-related left-ventricular dysfunction is multifactorial.15  After injection, daunorubicin is rapidly distributed to various tissues (eg, heart, lung, kidneys, spleen, liver, lean tissue). After cellular uptake, it is intercalated into mitochondrial and nuclear DNA. This causes double-strand breaks and activates topoisomerase-2β, which induces apoptosis and cellular death. It also causes mitochondrial dysfunction through the formation of reactive oxygen species (ROS) and endoplasmic reticulum (ER) stress. Other contributing mechanisms include titin proteolysis and inhibition of the neuregulin/ErbB pathway.15-17  All these processes contribute to the loss of functional cardiomyocytes, myocardial disarray, and development of interstitial fibrosis leading to left ventricular dysfunction and finally chronic heart failure.15,17,68  The histopathological pictures are a courtesy of Karin Klingel, Department of Cardiopathology, University Hospital Tübingen (Tübingen, Germany).

Close modal

There are several predisposing factors for ARLVD. The most important modifiable risk factor is the cumulative anthracycline dose (CAD). Other risk factors for ARLVD include female sex, age >65 or <18 years, renal failure, concomitant or previous radiation therapy involving the heart, concomitant chemotherapy with alkylating (eg, cyclophosphamide) or antimicrotubule agents (eg, taxanes), preexisting cardiac diseases such as systemic hypertension,18  and genetic factors (eg, the P450 oxidoreductase SNPs rs2868177, rs13240755, and rs4732513 are related to daunorubicin–induced cardiotoxicity19 ).

CAD

Chronic ARLVD is dependent on the CAD20  and correlates with peak plasma anthracycline concentrations.21 

Symptomatic HF does not generally develop with a cumulative (nonliposomal) doxorubicin dose of <400 mg/m2. At a cumulative dose of 400 to 550 mg/m2 doxorubicin, there is a 5% risk of developing HF, which increases to 25% to 43% at 700 mg/m2.4,18,20-22  Some studies suggest that other anthracycline analogs, such as epirubicin and idarubicin, have a lower risk of ARLVD relative to their therapeutic doses.23  Mitoxantrone, an anthracenedione, appears to have a higher risk of ARLVD in comparison with doxorubicin.24  In older adults, there is some evidence that the CAD needed to cause ARLVD is lower.4,25  This was shown by the pivotal study of von Hoff et al4  demonstrating higher incidences of ARLVD in adults >60 years even at lower CAD in comparison with younger adults (eg, incidence of ARLVD at a CAD of 250 mg/m2: 1.5% in adults aged 40-59 years vs 2.4% in adults aged ≥60 years; CAD of 400 mg/m2: 2.3% vs 4.6% and at 600 mg/m2 14.9% vs 22.4%).

A few studies suggest that ARLVD may develop at lower doses and may occur at a higher frequency than reported in the pivotal trials.3,26  Noteworthy, the data of and recommendations for maximum CAD are mostly derived from solid cancer and lymphoma regimens (Table 1) and may not be representative for the ARLVD risk in patients with AML undergoing induction therapy.27  Nonetheless, the incidence of ARLVD after AML induction therapy varies and was previously reported to be as high as 12% to 18% for standard “7+3” regimen.28-30  This is alarming considering that induction therapy generally comprises a cumulative daunorubicin dose of ∼180 to 360 mg/m2, whereas the expected CAD that leads to a >5% risk of ARLVD is between 400 and 550 mg/m2.20  This suggests that additional factors may contribute to the unexpected high incidence of ARLVD in AML, such as the relative dose density during induction therapy.31  In solid cancers, the CAD is usually given over several months, whereas in AML, anthracycline is given over a short period (eg, daunorubicin 60 mg/m2 on days 1-3). In addition, ARLVD was found to be more common in the setting of infections during AML therapy.30,31  Severe infections are more common during therapy for AML compared with other cancer types (eg, breast). Thus, infections may augment the higher risk of ARLVD in AML (Table 1).

Table 1.

Cumulative dose of different anthracycline agents that are associated with a >5% incidence of heart failure18,20-22 

Anthracycline agentCumulative dose, mg/m2*
Daunorubicin 400-550 
Doxorubicin 400-550 
Epirubicin 900 
Idarubicin (IV) 90 
Mitoxantrone 100-160 
Liposomal anthracyclines >1000 
Anthracycline agentCumulative dose, mg/m2*
Daunorubicin 400-550 
Doxorubicin 400-550 
Epirubicin 900 
Idarubicin (IV) 90 
Mitoxantrone 100-160 
Liposomal anthracyclines >1000 
*

Data on cumulative doses associated with risk of heart failure are mostly derived from trials of patients with breast cancer, sarcoma, and lymphoma. Although the dose density and schedules are different in leukemia therapies, the cumulative dose for anthracycline-related toxicity is applicable in other cancer types.

Data for mitoxantrone are mixed; reported is an ARLVD incidence of 2.6% at a cumulative dose of 140 mg/m2 with the strict recommendation not to exceed this dose.121 

Other risk factors for ARLVD

Pretreatment comorbidities such as renal failure, hypertension, and preexisting cardiac diseases are common in the older adult population (Table 2) and are concomitant risk factors for the development of ARLVD.

Table 2.

Prevalence of risk factors for anthracycline-related left ventricular dysfunction in the general older adult population

ComorbidityAge, yPrevalence in the general older adult population, %
Arterial hypertension122  >60 60-70 
Chronic heart failure*123-125  60-70 ∼4-8 
70-79 9-12 
80+ >15 
Coronary artery disease126  >60 15-36 
Peripheral artery disease127  60-70 7.1 
71-80 13 
80+ >22 
Chronic renal insufficiency128,129  60-69 ∼9-13 
70+ 11-46 
ComorbidityAge, yPrevalence in the general older adult population, %
Arterial hypertension122  >60 60-70 
Chronic heart failure*123-125  60-70 ∼4-8 
70-79 9-12 
80+ >15 
Coronary artery disease126  >60 15-36 
Peripheral artery disease127  60-70 7.1 
71-80 13 
80+ >22 
Chronic renal insufficiency128,129  60-69 ∼9-13 
70+ 11-46 
*

Including asymptomatic left ventricular ejection fraction decline, excluding those with preserved ejection fraction

Glomerular filtration rate <60 mL/min/1.73 m2.

In the broader context, frailty is a well-known risk factor for increased chemotherapy toxicities.32  Frailty is defined as “a clinical state in which there is an increase in an individual’s vulnerability for developing increased dependency and/or mortality when exposed to a stressor.”33  Frailty is more common in the geriatric population and is associated with sarcopenia, sarcopenic obesity, and hypoalbuminemia, all of which could lead to changes in pharmacokinetics of anthracyclines. Nonetheless, data on changes in pharmacokinetics of anthracycline with age are conflicting.34-36  Daunorubicin has a high distribution volume, intensive tissue uptake, and moderate plasma protein binding.36,37  Therefore, it is possible that a change in body composition may decrease tissue distribution leading to higher peak concentration resulting in higher risk of ARLVD.38  In animal models, higher peak concentrations of daunorubicin were found in older rats compared with young rats.39  In another study using a rat and rabbit model, protein malnutrition, lower body weight, and hypoalbuminemia alter pharmacokinetics of doxorubicin with a significant decrease in drug elimination.39,40  It is unclear whether similar findings are seen in patients who are cachectic or sarcopenic but the findings in animal models could provide an explanation for the increased toxicities observed in older adults.

Preexisting cardiac comorbidities are common reasons for excluding older adults from receiving intensive chemotherapy.41,42  An analysis of the Danish National Leukemia Registry demonstrated that either heart or renal failure are common reasons why intensive therapy was not given.41  Even in patients ≥60 years selected for intensive induction therapy based on their overall condition, prevalence of cardiac comorbidities is high.41,43-46 Table 3 shows the prevalence of cardiac and related comorbidities in patients with AML. Cardiovascular comorbidities are poor prognostic factors and are reflected by the Hematopoietic Stem-Cell Transplantation–Comorbidity Index (HCT-CI), which is a validated tool to predict mortality in patients with AML and those undergoing allogeneic hematopoietic stem cell transplantation (HSCT).43,47  In this tool, several cardiac comorbidities (eg, coronary artery disease, HF, arrhythmias, heart valve abnormalities) are shown to be associated with poor outcomes.

Table 3.

Prevalence of risk factors for anthracycline-related left ventricular dysfunction in patients with AML

AML cohortAge groupComorbidityPrevalence, %
Danish National Leukemia Registry*41  Median age 59 y (range: 15-83 y) Myocardial infarction 24.6 
Chronic heart failure 12.5 
Peripheral vascular disease 21.9 
Moderate to severe renal disease 19.2 
MD Anderson Cancer Center (HCT-CI validation cohort)*45  >60 y Cardiac disease (chronic heart failure, coronary artery disease, valvular dysfunction) 41.0 
Prior cerebrovascular accident 6.0 
Comprehensive Cancer Center of Wake Forrest University*44  ≥60 y Cardiac disease 12.5 
Renal disease 15.3 
Surveillance, Epidemiology, and End-Results Medicare linked database45  >65 y; mean age 77.26 y ± 6.96 Chronic heart failure 21.5 
Cerebrovascular disease 18.2 
Prior myocardial infarction 8.2 
AML cohortAge groupComorbidityPrevalence, %
Danish National Leukemia Registry*41  Median age 59 y (range: 15-83 y) Myocardial infarction 24.6 
Chronic heart failure 12.5 
Peripheral vascular disease 21.9 
Moderate to severe renal disease 19.2 
MD Anderson Cancer Center (HCT-CI validation cohort)*45  >60 y Cardiac disease (chronic heart failure, coronary artery disease, valvular dysfunction) 41.0 
Prior cerebrovascular accident 6.0 
Comprehensive Cancer Center of Wake Forrest University*44  ≥60 y Cardiac disease 12.5 
Renal disease 15.3 
Surveillance, Epidemiology, and End-Results Medicare linked database45  >65 y; mean age 77.26 y ± 6.96 Chronic heart failure 21.5 
Cerebrovascular disease 18.2 
Prior myocardial infarction 8.2 
*

Includes only patients preselected to receive intensive chemotherapy.

As defined by Charlson Comorbidity Index (chronic heart failure defined by symptomatic dyspnea; chronic renal disease defined as creatinine >3 mg/dL).

Based on International Classification of Diseases-9- and Current Procedural Terminology-4 codes.

The effect of ARLVD on outcomes was assessed in a large cohort of pediatric AML patients within the Children’s Oncology Group trial AAML0531. ARLVD was associated with a reduction in event-free survival (EFS) and overall survival (OS).30  Although the study was performed in the pediatric population, it reinforces the negative prognostic implications of ARLVD. A recent single-center retrospective analysis also demonstrated a reduction in OS after HSCT in the setting of ARLVD (including a transient LVEF decline) that developed after induction therapy (13 vs 27 months; P = .013).29  Because a curative treatment approach may include an HSCT in select patients, a transient LVEF decline may potentially delay HSCT, thereby worsening outcomes.

When considering the use of anthracycline, it is also important to consider leukemia-related factors that are associated with a poor response to “7+3” regimen. In this case, the risk for ARLVD may exceed the potential benefit derived from anthracycline-based regimen. Factors that are related to chemoresistance include TP53 mutations, complex cytogenetic aberrations,48  and increased expression of multidrug-resistance proteins such as p-glycoprotein–based efflux pumps.49  In contrast, NPM1 mutations or core-binding factor leukemia are predictive of chemosensitive disease. Prognostic and predictive factors are summarized in Table 4.

Table 4.

Response rates to “7+3”-like regimens according to molecular and cytogenetic risk groups in older adults

SourceStudy populationCR 1, %Long-term survival, %
Vasu et al, 2019130  944 pts with de novo AML >70 y, enrolled on intensive therapy CALGB/Alliance protocols without HSCT  At 10 y: 
CBF AML ∼92 ∼17 
Cytogenetically normal AML ∼68 ∼2.4 
Abnormal karyotype AML ∼51 ∼1.1 
Prebet et al, 2009131  CBF leukemia >60 y, received intensive induction therapy 88 At 5 y: 31 
Eisfeld et al, 2018132  423 pts with de novo AML age 60-85 y having received intensive induction therapy without HSCT in CR  At 3 y: 
ELN 2017 favorable risk 82 30 
ELN 2017 intermediate risk 54 12 
ELN 2017 unfavorable risk 38 
NPM1mut + IDH2/SF1/SRSF2mut NA 45 
FLT3-ITDmut/TP53mut/BCORmut/U2AF1mut/WT1mut/complex karyotype/t(9;11) NA 
Ostronoff et al, 2015133  Pts ≥55years with NPM1+ AML, received intensive induction therapy  At 2 y: 
55-65 y 67 39 
>65 y 53 19 
SourceStudy populationCR 1, %Long-term survival, %
Vasu et al, 2019130  944 pts with de novo AML >70 y, enrolled on intensive therapy CALGB/Alliance protocols without HSCT  At 10 y: 
CBF AML ∼92 ∼17 
Cytogenetically normal AML ∼68 ∼2.4 
Abnormal karyotype AML ∼51 ∼1.1 
Prebet et al, 2009131  CBF leukemia >60 y, received intensive induction therapy 88 At 5 y: 31 
Eisfeld et al, 2018132  423 pts with de novo AML age 60-85 y having received intensive induction therapy without HSCT in CR  At 3 y: 
ELN 2017 favorable risk 82 30 
ELN 2017 intermediate risk 54 12 
ELN 2017 unfavorable risk 38 
NPM1mut + IDH2/SF1/SRSF2mut NA 45 
FLT3-ITDmut/TP53mut/BCORmut/U2AF1mut/WT1mut/complex karyotype/t(9;11) NA 
Ostronoff et al, 2015133  Pts ≥55years with NPM1+ AML, received intensive induction therapy  At 2 y: 
55-65 y 67 39 
>65 y 53 19 

CBF, core binding factor; CR 1, first complete remission; ELN 2017, European Leukemia Net risk stratification 2017; mut, mutated; NA, not addressed; pts, patients.

The integration of cardio-oncology teams appears to be of utmost importance. A recent analysis of AML patients with ARLVD found that only a minority was seen by a cardiologist and received HF medication.50  In a Canadian survey, <20% of hematologists stated that they regularly consider the indication before they stop cardiovascular medication in AML patients and up to 28% did not restart this medication after interruption.51  This clearly illustrates that cardio-oncology specialists might better assess for risk factors of ARLVD and recommend preventive and therapeutic strategies. Nonetheless, cardio-oncologists are not broadly available. For routine practice, we recommend partnerships with cardiologists with experience in managing ARLVD to jointly follow older adults receiving anthracycline-based induction therapy who have risk factors for ARLVD.

Although there are several guidelines in the assessment and surveillance of adults receiving anthracycline-based chemotherapy,18,52,53  data specific to older adults with AML are limited. Strategies for assessing cardiac abnormalities include cardiac imaging (echocardiography, nuclear imaging, magnetic resonance imaging) and biomarkers (troponin, natriuretic peptides).18 

Transthoracic echocardiography is a widely available and noninvasive modality to assess cardiac function. Although multiple-gated acquisition is also commonly used, it involves radiation exposure and is not able to assess atrial pressures, right ventricular, and valvular functions.54  Cardiac magnetic resonance imaging (CMRI) can be used to assess cardiac function, especially when other noninvasive imaging is inconclusive. Although the availability of CMRI may be more limited in certain health care situations compared with other modalities, CMRI has been suggested to be more sensitive at detecting asymptomatic ARLVD than echocardiography in long-term follow-up55  and very early changes after treatment.56 

Serial monitoring of LVEF by repetitive transthoracic echocardiography is recommended before, during, and after anthracycline treatment to monitor for ARLVD without consensus for timing.12,18,52,53  However, measuring only LVEF detects changes that may already be irreversible at the time of diagnosis. Advanced imaging techniques such as echocardiography-derived measures of global longitudinal strain and CMRI may detect early stages of cardiac dysfunction and predict future ARLVD.57,58  For detailed recommendations on imaging, please refer to the joint American and European recommendations.12 

Cardiac biomarkers are currently being investigated for their role in predicting ARLVD and to identify patients who could benefit from cardioprotective therapy.59  Studies involving adults receiving high-dose chemotherapy have linked early increases in troponin to subsequent ARLVD.60,61  However, in these trials, troponin was mostly determined weeks after prior anthracycline application, which was given over several treatment cycles. In AML, anthracyclines are usually given in a dose-dense manner (eg, days 1-3/3-5 of chemotherapy) and not over repetitive cycles of treatment. In addition, troponin may be elevated due to acute change in clinical status (eg, hyperleukocytosis,62  pneumonia63 ) but still indicates the extent of related myocardial damage. Despite its promising role as predictive biomarker, there are currently not enough data to support a cardiac risk stratification in AML patients based on troponin measurements, and more research is warranted.

Several strategies have been investigated to minimize ARLVD as discussed in the following section.

Chelation therapy (eg, dexrazoxane)

Dexrazoxane is approved by the US Federal Drug Administration for prevention of cardiotoxicity in patients with breast cancer who have reached a CAD of 300 mg/m2 and require additional doxorubicin.64,65  In adult patients with AML, 3 case series on the use of dexrazoxane have been published to date (N = 15 patients).66-68  All patients aged ≥60 years (N = 4) died of various reasons soon after treatment. Despite the lack of solid data on dexrazoxane use in older adults with AML, its cardioprotective properties and its safety profile in childhood leukemia are promising.69  Dexrazoxane should be further evaluated in larger randomized trials of older individuals.

Prolonged anthracycline infusion time

High peak plasma levels of anthracyclines are associated with ARLVD. Prolonged infusion time reduces the peak plasma levels and was therefore investigated for its potential to reduce ARLVD.70  A Cochrane meta-analysis compared an infusion duration of 6 hours or longer with a shorter duration of anthracycline doses equivalent to 30 to 60 mg/m2 doxorubicin.71  The risk of clinical HF was lower (risk ratio, 0.27; 95% confidence interval, 0.09-0.81).71  Potentially, it decreases also asymptomatic LVEF decline.71  Thus, a prolonged infusion time can be considered for individuals at risk.

Use of liposomal formulations

Liposomal formulations of anthracyclines have a prolonged plasma half-time and decreased distribution volumes compared with nonliposomal formulation.72  This potentially reduces the likelihood of ARLVD.72,73  There are currently pegylated and nonpegylated liposomal doxorubicin as well as liposomal daunorubicin and dual-drug liposomal formulation of cytarabine and daunorubicin (CPX-351).

A randomized phase 2 trial (GIMEMA GSI 103 AMLE) assessed the efficacy between liposomal daunorubicin (80 mg/m2 on days 1-3) vs standard daunorubicin (45 mg/m2 on days 1-3) in patients aged ≥60 years during AML induction.74  There were no differences in complete remission (CR) rates, or ARLVD between the groups. Therefore, liposomal daunorubicin is not commonly used.

CPX-351 is approved by the US Federal Drug Administration for upfront treatment of patients with therapy-related AML, or AML with myelodysplasia-related changes.75  It is a dual-drug liposomal formulation of cytarabine and daunorubicin delivered in a fixed 5:1 molar ratio. In vitro studies demonstrated that the maximal synergy is achieved by the fixed 5:1 concentration of cytarabine and daunorubicin. Furthermore, because cellular uptake of CPX-351 is mainly achieved via liposomes, P-glycoprotein–based efflux pumps are likely bypassed. The latter is a common cause of anthracycline-resistance and also occurs more often in older adult.76  The initial phase 1 study77  excluded patients with LVEF <50%. Among the 48 patients, 23 had both pre- and posttherapy LVEF data available; 12 patients received cumulative daunorubicin dose of >400 mg/m2. Of those, symptomatic LVEF decline was noted in 2 patients. In the phase 3 study comparing CPX-351 with “7+3” regimen, no significant differences in ARLVD were noted between the 2 arms.75  At this point in time, it is uncertain if CPX-351 has a lower risk of ARLVD, but we do not consider CPX-351 as less cardiotoxic so far.

Alternative daunorubicin dosing

Although it is possible that a lower CAD may be sufficient to cause ARLVD, the optimal anthracycline dose for induction therapy is still a matter of debate.3,4,29  Interestingly, a dose reduction of doxorubicin and cyclophosphamide within the rituximab, cyclophosphamide, doxorubicin hydrochloride, vincristine sulfate, and prednisone regimen in adults ≥80 years with aggressive lymphoma showed a similar efficacy in comparison with the full dose in younger patients.78  This raises the question whether the effective dose of anthracycline is different in older adults because of changes in body composition and whether the standard dose of daunorubicin (60-90 mg/m2) is needed to achieve an equivalent efficacy.

Three trials have assessed the efficacy of different daunorubicin dosing comparing daunorubicin 35 mg/m2 with 50 mg/m2,79  60 mg/m2 to 90 mg/m2,80  and 45 mg/m2 to 90 mg/m2,81  without any significant impact on outcome. None of these trials included patients with a reduced LVEF, or monitored LVEF longitudinally. Therefore, an impact on subclinical and long-term cardiac function could not be specified.

Based on these studies, it appears that a lower dose of anthracycline may be as effective as standard dosing in older patients with AML. Although these studies generally excluded patients with cardiac risk factors, it may be worth considering a lower dose of anthracycline in older patients with AML, particularly those with cardiac risk factors, although this remains an individual decision.

HF medication as preventive and early therapeutic strategy

Angiotensin-converting enzyme inhibitors (ACE-Is) and β-blockers are frequently used for management of LVEF decline and HF, and have been shown to reduce mortality in older adults with HF.82-87  ACE-Is have also been shown to delay the progression of left ventricular dysfunctions, including ARLVD.88 

Table 5 summarizes various clinical studies evaluating primary and secondary preventive strategies using β-blockers and/or ACE-Is for ARLVD. The results are heterogeneous and are likely the result of different study populations and CAD. Only 1 study (Prevention of Left Ventricular Dysfunction with Enalapril and Carvedilol in Patients Submitted to Intensive Chemotherapy for the Treatment of Malignant Hemopathies trial31 ) evaluated a primary preventive strategy in a larger group of patients with acute leukemia (among other cancer types). Compared with the patients with AML in the control arm who had a significant absolute decrease in LVEF of 6.4%, patients in the treatment arm who received enalapril and carvedilol had preserved LVEF. Patients with preexisting LVEF <50% were excluded from the study. In addition, the CAD was low (mean CAD <300 mg/m2). Although the difference between the 2 groups was significant, only a small number of patients had their LVEF drop below 50%; most of the patients technically did not fulfill the criteria of ARLVD despite the statistically significant LVEF drop. Remarkably, in the intention-to-treat analysis, the combined endpoint of death, HF, or LVEF <45% occurred in 24.4% in the control group vs 6.7% in the intervention group.31 

Table 5.

Overview of clinical trials that assess the efficacy of an ACE-I and/or a β-blocker for primary and secondary prevention of anthracycline-related left ventricular dysfunction

StudyTrial populationInterventionEndpoints of interestOutcomeFindings
Primary preventive strategies and biomarker-triggered primary prevention 
Bosch et al, 201331  Acute leukemia, autologous HSCT; mean age 50 ± 13 y, LVEF >50% prechemotherapy Enalapril and carvedilol vs no preventive medication (control group) Death, HF, or a final LVEF decline to <45% Death and HF 6.7% (enalapril/carvedilol group) vs 22.2% (control group), P = .02; prespecified subgroup analysis in leukemia pts: no LVEF change in enalapril/carvedilol group, 6.7% absolute decrease in control group (P = .025) Subgroup analysis of leukemia pts included 
OVERCOME Trial 
Georgakopoulos et al, 2010134  Hodgkin lymphoma and NHL; mean age 49 y; ACC Metoprolol vs enalapril vs no preventive medication LVEF decline Enalapril and metoprolol did not affect the probability of developing heart failure Exclusively lymphoma pts 
Kalay et al, 2006135  Mainly BC and lymphoma patients, mean age 46.8 ± 14 y (carvedilol group) and 49.0 ± 9.8 y (control group); ACC Carvedilol vs no preventive medication LVEF decline Significantly lower LVEF and higher incidence of systolic dysfunction in control group Small patient numbers (25+25 pts). No AML pts included as explicitly stated 
Kaya et, 2013136  BC; mean age 51.4 ± 9.4 y (nebivolol group) and 50.5 ± 11.1 y (control group); ACC Nebivolol vs no preventive medication LVEF decline, NT-proBNP increase LVESD/LVEDD increase and LVEF decline in control group, increase of NT-proBNP; no significant changes in nebivolol group Small patient numbers (45 pts). No AML included 
Gulati et al, 2017137  BC; mean age not specified (mostly <60 y); ACC Metoprolol vs candesartan vs metoprolol and candesartan vs placebo LVEF decline, increase in cardiac biomarkers Metoprolol attenuates troponin rise; no effect for candesartan; significant LVEF decline in placebo group Exclusively BC pts 
PRADA Trial 
Cardinale et al, 201892  Various cancers; mean age 51 ± 12 y; ACC Enalapril preventive or triggered by troponin-rise Troponin rise; LVEF decline No attenuation of troponin rise with enalapril; LVEF decline negligible in both arms ACC with low cumulative doses ± trastuzumab and/or taxane, few acute leukemia pts included 
ICOS-ONE Trial 
Cardinale et al, 200691  Various cancers; inclusion after early TnI rise; mean age 47 ± 11 y (enalapril) and 47 ± 13 y (control), various chemotherapy schemes and partially ACC  Enalapril vs no preventive medication (control group) LVEF decline ≥10% of baseline below 50% LVEF decline >10% in 0% (enalapril group) vs 43% (control group) Few leukemia pts included 
Secondary preventive/therapeutic strategies 
Cardinale et al, 201510  Mainly BC and NHL, inclusion after drop of LVEF >10%; mean age 50 ± 13 y, ACC Enalapril ± β blockers started after LVEF drop LVEF recovery 82% recovered from ARC No randomization for HF therapy; no AML pts included as explicitly stated 
Cardinale et al, 201093  Various cancers; inclusion after LVEF drop to ≤45% (asymptomatic and symptomatic); mean age 53 ± 12 y; ACC Enalapril ± carvedilol LVEF recovery 42% responders (normalization of LVEF in 42% and partial recovery in 13%). No AML pts included 
0% recovery if HF therapy started >6 mo after ACC, frequent adverse cardiac events in nonresponders 
Silber et al, 2004138  Pediatric cancer long-term survivors; LVEF drop several years after ACC Enalapril vs placebo Exercise tolerance, LVESWS, LVEF recovery No change in exercise tolerance or LVEF Late start of HF therapy 
AAA Trial LVESWS significantly reduced within the first treatment year 
StudyTrial populationInterventionEndpoints of interestOutcomeFindings
Primary preventive strategies and biomarker-triggered primary prevention 
Bosch et al, 201331  Acute leukemia, autologous HSCT; mean age 50 ± 13 y, LVEF >50% prechemotherapy Enalapril and carvedilol vs no preventive medication (control group) Death, HF, or a final LVEF decline to <45% Death and HF 6.7% (enalapril/carvedilol group) vs 22.2% (control group), P = .02; prespecified subgroup analysis in leukemia pts: no LVEF change in enalapril/carvedilol group, 6.7% absolute decrease in control group (P = .025) Subgroup analysis of leukemia pts included 
OVERCOME Trial 
Georgakopoulos et al, 2010134  Hodgkin lymphoma and NHL; mean age 49 y; ACC Metoprolol vs enalapril vs no preventive medication LVEF decline Enalapril and metoprolol did not affect the probability of developing heart failure Exclusively lymphoma pts 
Kalay et al, 2006135  Mainly BC and lymphoma patients, mean age 46.8 ± 14 y (carvedilol group) and 49.0 ± 9.8 y (control group); ACC Carvedilol vs no preventive medication LVEF decline Significantly lower LVEF and higher incidence of systolic dysfunction in control group Small patient numbers (25+25 pts). No AML pts included as explicitly stated 
Kaya et, 2013136  BC; mean age 51.4 ± 9.4 y (nebivolol group) and 50.5 ± 11.1 y (control group); ACC Nebivolol vs no preventive medication LVEF decline, NT-proBNP increase LVESD/LVEDD increase and LVEF decline in control group, increase of NT-proBNP; no significant changes in nebivolol group Small patient numbers (45 pts). No AML included 
Gulati et al, 2017137  BC; mean age not specified (mostly <60 y); ACC Metoprolol vs candesartan vs metoprolol and candesartan vs placebo LVEF decline, increase in cardiac biomarkers Metoprolol attenuates troponin rise; no effect for candesartan; significant LVEF decline in placebo group Exclusively BC pts 
PRADA Trial 
Cardinale et al, 201892  Various cancers; mean age 51 ± 12 y; ACC Enalapril preventive or triggered by troponin-rise Troponin rise; LVEF decline No attenuation of troponin rise with enalapril; LVEF decline negligible in both arms ACC with low cumulative doses ± trastuzumab and/or taxane, few acute leukemia pts included 
ICOS-ONE Trial 
Cardinale et al, 200691  Various cancers; inclusion after early TnI rise; mean age 47 ± 11 y (enalapril) and 47 ± 13 y (control), various chemotherapy schemes and partially ACC  Enalapril vs no preventive medication (control group) LVEF decline ≥10% of baseline below 50% LVEF decline >10% in 0% (enalapril group) vs 43% (control group) Few leukemia pts included 
Secondary preventive/therapeutic strategies 
Cardinale et al, 201510  Mainly BC and NHL, inclusion after drop of LVEF >10%; mean age 50 ± 13 y, ACC Enalapril ± β blockers started after LVEF drop LVEF recovery 82% recovered from ARC No randomization for HF therapy; no AML pts included as explicitly stated 
Cardinale et al, 201093  Various cancers; inclusion after LVEF drop to ≤45% (asymptomatic and symptomatic); mean age 53 ± 12 y; ACC Enalapril ± carvedilol LVEF recovery 42% responders (normalization of LVEF in 42% and partial recovery in 13%). No AML pts included 
0% recovery if HF therapy started >6 mo after ACC, frequent adverse cardiac events in nonresponders 
Silber et al, 2004138  Pediatric cancer long-term survivors; LVEF drop several years after ACC Enalapril vs placebo Exercise tolerance, LVESWS, LVEF recovery No change in exercise tolerance or LVEF Late start of HF therapy 
AAA Trial LVESWS significantly reduced within the first treatment year 

ACC, anthracycline-containing chemotherapy; ARC, anthracycline-related cardiotoxicity; BC, breast cancer; LVESD, left ventricular end-systolic diameter; LVEDD, left ventricular end diastolic diameter; LVESWS, left ventricular end-systolic wall stress; NHL, non-Hodgkin lymphoma; TnI, troponin I.

Interestingly, more patients in the control group died of infection compared with the treatment group. Furthermore, those who had sepsis and survived had a significant reduction in their LVEF. Based on these findings, the authors speculated that the use of ACE-Is as primary prevention may influence outcomes of patients who experience infections.31  This may be related to the inhibition of the renin-angiotensin-aldosterone system that, when activated, promotes inflammation and endothelial damage during sepsis.89  Prior study has demonstrated that prehospital ACE-Is in the general population may improve outcome in patients with septic shock.90 

In addition to the primary preventive strategy, biomarker-triggered strategies such as initiating enalapril after detection of increased troponin levels have also been found to be effective in a randomized controlled trial in different cancer entities.91,92  Nonetheless, it is unclear when and how frequent troponin should be measured, given that elevated troponin may be not anthracycline-related in the acute setting.

None of the aforementioned studies included patients aged >70 years and those aged 60 to 70 years were underrepresented.10,31,92,93  Therefore, there is low-certainty evidence for benefit in the older population. However, ACE-Is are generally well tolerated in older adults,94  with usual consideration given to the risk of adverse reaction (eg, hyperkalemia) in the presence of comorbidities (eg, renal insufficiency). Given the evidence for use of ACE-Is, this may be considered with or without the use of a β-blocker as a therapy for use during receipt of anthracyclines when early left ventricular dysfunction occurs.

Considerable numbers of older patients with AML present with already declined LVEF or cardiac comorbidity rendering the use of anthracycline containing chemotherapy regimens hazardous. Other than preventive strategies for ARLVD, several efforts have been used to avoid anthracycline use. Here, we discuss alternative treatment strategies.

No randomized data are available that compare non-anthracycline induction strategies in the presence of impaired LVEF. However, expert opinion suggests that non-anthracycline induction strategies should be considered where there is concern for tolerability of anthracycline-containing induction therapy.95  Several anthracycline-free intensive induction regimens have shown similar efficacy in comparison with “7+3” regimen, although only a minority was tested in randomized trials.96-100  These options are listed in Table 6.

Table 6.

Overview of clinical trials that assess anthracycline-free induction strategies

StudyTrial populationInterventionTrial designOutcome
Vulaj et al, 2018 97  Secondary AML, mean age 63 y (range, 27-82) in the FLAG group (N = 40), 60 y (range, 21-76) in the 7+3 group (N = 66); no data on LVEF provided 7+3 vs FLAG Retrospective single-center analysis Higher RR in the FLAG group vs 7+3 (70% vs 48%, P = .043), no significant difference in OS (8.5 vs 9.1 mo; P = .798), shorter duration of neutropenia in the FLAG group (16 vs 23 d, P < .001) 
FOSSIL Study 
Bashey et al, 2006 96  AML, aged >60 y; no previous therapy (N = 24), no data on LVEF provided FLAG Retrospective single-center analysis CR rate 58%, 17% CRI 
Brunnberg et al, 201298  AML, ≥60 y; previously untreated (N = 119), exclusion of patients with HF NYHA III-IV “7+3” vs “7+GO” Randomized phase 2 trial Both regimens were equally effective in blast clearance, CR, EFS, remission duration, and OS. Higher induction death rate from veno-occlusive disease in the “7+GO” group. No cardiac toxicities in both groups were observed 
Kessler et al, 200899  AML ≥60 y; TAA (N = 16) vs TAD/HAM (N = 16) TAD/HAM vs TAA in the setting of HF (NYHA III-IV or LVEF <40%) Single-center, matched-pair analysis No significant difference in RR, RFS, and OS 
Borthakur et al, 2008100  CBF-AML (age range 16-83 y) FLAG vs FA vs IA ±Retrospective single-center analysis No significant difference in RFS and OS between the groups 
StudyTrial populationInterventionTrial designOutcome
Vulaj et al, 2018 97  Secondary AML, mean age 63 y (range, 27-82) in the FLAG group (N = 40), 60 y (range, 21-76) in the 7+3 group (N = 66); no data on LVEF provided 7+3 vs FLAG Retrospective single-center analysis Higher RR in the FLAG group vs 7+3 (70% vs 48%, P = .043), no significant difference in OS (8.5 vs 9.1 mo; P = .798), shorter duration of neutropenia in the FLAG group (16 vs 23 d, P < .001) 
FOSSIL Study 
Bashey et al, 2006 96  AML, aged >60 y; no previous therapy (N = 24), no data on LVEF provided FLAG Retrospective single-center analysis CR rate 58%, 17% CRI 
Brunnberg et al, 201298  AML, ≥60 y; previously untreated (N = 119), exclusion of patients with HF NYHA III-IV “7+3” vs “7+GO” Randomized phase 2 trial Both regimens were equally effective in blast clearance, CR, EFS, remission duration, and OS. Higher induction death rate from veno-occlusive disease in the “7+GO” group. No cardiac toxicities in both groups were observed 
Kessler et al, 200899  AML ≥60 y; TAA (N = 16) vs TAD/HAM (N = 16) TAD/HAM vs TAA in the setting of HF (NYHA III-IV or LVEF <40%) Single-center, matched-pair analysis No significant difference in RR, RFS, and OS 
Borthakur et al, 2008100  CBF-AML (age range 16-83 y) FLAG vs FA vs IA ±Retrospective single-center analysis No significant difference in RFS and OS between the groups 

CBF-AML, core binding factor AML; CRI, complete remission with incomplete count recovery; EFS, event-free survival; FLAG, fludarabine 30 mg/m2 per day on days 1-5, cytarabine 2 g/m2 per day on days 1-5, filgrastim 300-480 µg/d on day 1-count recovery; HAM, cytarabine 1 g/m2 every 12 hours on days 1-3, mitoxantrone 10 mg/m2 on days 3-5; IA ± filgrastim, idarubicin 12 mg/m2 on days 1-3, cytarabine 1.5 g/m2 per day p.c. on days 1-4 (1-3 if age >65 years), ± filgrastim 5 µg/kg on days −1 to 5; NYHA, New York Heart Association, 7+GO, standard cytarabine 100 mg/m2 per day days 1-7, gemtuzumab-ozogamicin 6 mg/m2 on day 1, 4 mg on day 8; RFS, relapse-free survival; RR, response rate; TAA, 6-thioguanine 200 mg/m2 on days 3-9, cytarabine 100 mg/m2per 24 hours every 12 hours on days 1-2, and 100 mg/m2 on days 3-8, Amsacrine 210 mg/m2 on days 3-5; TAD, 6-thioguanine 200 mg/m2 on days 3-9, cytarabine 100 mg/m2 per 24 hours on days 1-2 or 100 mg/m2, every 12 hours on days 3-8, and daunorubicin 60 mg/m2 on days 3-5.

Other non-anthracycline strategies include treatments such as hypomethylating agents (HMA [eg, decitabine, 5-azacytidine], BCL-2 inhibitors [eg, venetoclax], FLT3 or IDH inhibitors). A randomized controlled study suggests similar survival rates using HMA compared with intensive induction therapy.101  Furthermore, certain high-risk subgroups may benefit from HMA such as those with TP53 mutations, or complex karyotypes.102-104  A head-to-head comparison between decitabine and “3+7” as the preferred induction strategy for older patients is currently being tested in the InDACtion trial.105 

The BCL-2 inhibitor venetoclax in combination with HMA106  or low-dose cytarabine107  has shown promising results. The phase 1b study106  that evaluated HMA and venetoclax demonstrated a CR and complete remission with incomplete recovery rate of 73% with an overall response rate of 73% (comprising CR, incomplete recovery, and partial remission).106 

Targeted therapeutics, such as FLT3 or IDH inhibitors, represent another promising strategy for this patient group, also in novel combinations with HMAs. The FLT3 inhibitor sorafenib showed favorable risk ratios in combination with azacytidine in AML patients aged 61 to 86 years.108  Of note, although sorafenib harbors a known risk for LVEF decline and adverse cardiovascular events,18  none of the landmark trials for sorafenib use in AML, including those in older adults, showed a significantly increase of these adverse events. Nonetheless, the small sample sizes in these studies limited our ability to draw definite conclusions.108-111 

IDH inhibitors (ivosidenib and enasidenib) carry the risks of QTc prolongation without any other known cardiotoxicity.112,113  These agents are being explored in the upfront setting. They could be potential upfront therapeutic options for patients with IDH mutations with cardiac risk factors that preclude the use of anthracycline-based regimens.

These combination strategies will likely replace single-agent therapy alone and appear to be reasonable options for patients not candidates for anthracycline-containing induction therapy, also as a possible bridging strategy to HSCT.106,114 

A baseline LVEF ≤50% is most commonly used as an exclusion criterion in clinical trials containing an anthracycline.81,115  Outside clinical trials, this threshold might differ depending on the center and physician’s preferences. Often, there is a concern that anthracycline use in patients with mild LVEF decline could lead to acute HF. However, an asymptomatic HF with mid-range reduced ejection fraction (LVEF, 40% to 49%) should not abrogate a curative approach if the patient is otherwise regarded as fit for intensive therapy and the cardiac comorbidity is well-controlled.95  This is especially true for patients with a favorable-risk AML such as those with NPM1 mutations or CBF aberrations. If a curative approach includes an HSCT, a history of cardiac comorbidity increases the risk of nonrelapse mortality as predicted by the surrogate assessment tool HCT-CI.47,116  HSCT in patients with decreased LVEF in the range of HF–mid-range reduced ejection fraction is probably feasible.117-119  Therefore, patients who are not eligible for anthracycline-based chemotherapy may still be able to receive HSCT. A patient judged as “unfit for anthracycline and fit for HSCT” could benefit from an alternative induction strategy, as described previously. Nonetheless, this remains an individualized decision, taking account into patient preference and goals. In patients who are less likely to respond to anthracycline-containing therapy, such as patients with TP53 mutation, we encourage consideration of non-anthracycline-containing therapy as the potential risk for ARLVD exceeds the potential benefit.

Emerging studies have suggested the role for advanced imaging techniques and biomarkers in early detection of ARLVD. However, there is a lack of consensus on their application in routine cardiac surveillance. Large, prospective, multi-institutional studies are needed to determine whether these imaging techniques and biomarkers allow an improved early detection of ARLVD, predict cardiovascular and OS, and inform early intervention strategies to reduce cardiovascular morbidity.120  Most patients develop ARLVD within the first year after therapy,10,29  and the recovery rate decreases rigorously after 6 months.10  Therefore, close cardiac monitoring is important. In case of ARLVD, the standard therapy for HF including ACE-Is, β-blocker, and eventually mineralocorticoid receptor antagonist should be started promptly.93  In case of ongoing therapy, a rigorous fluid management should be provided together with a close cardiac monitoring. Because a substantial number of patients develop ARLVD after completion of therapy, we recommend a continued comanagement with a cardiologist to optimize the cardiovascular risk profile.

In conclusion, older adults with AML are at risk of experiencing ARLVD. Based on our review, we summarized our recommendations in Table 7. There is high-certainty evidence to support the need to consider lifetime CAD and for serial monitoring for ARLVD. Several preventive strategies have been evaluated including alternative dosing, use of cardioprotective medications, liposomal formulation, and alternative induction strategies. More high-quality randomized controlled trials are required to elucidate the effectiveness of strategies in the prevention and treatment of ARLVD in older adults with AML.

Table 7.

Recommendations for assessment, alternative strategies, and management of anthracycline-related left ventricular dysfunction in older adults with AML

Baseline assessment 
 • Assess for preexisting cardiac comorbidities, risk factors for anthracycline-related left ventricular dysfunction, and frailty before therapy. 
 • Further assessment of clinical signs of HF and cardiac echocardiography should be performed before, during, and after therapy to evaluate for LVEF and if available, global longitudinal strain. 
Prevention 
 • If anthracycline can be administered safely with a prolonged infusion time, this should be considered, especially in patients with preexisting risk factors for anthracycline-related left ventricular dysfunction besides age, or a HF with mid-range reduced ejection fraction. 
Choice of induction regimens according to disease biology and risk factors for anthracycline-related left ventricular dysfunction 
 • In patients with chemotherapy-sensitive leukemia (eg, core-binding factor leukemia, or NPM1-mutated AML), a mild decrease in LVEF should not abrogate a curative approach and standard “7+3” regimen should be considered with the previously mentioned preventive strategy. Other options include reducing daunorubicin dose to 45 mg/m2 or use alternative intensive induction strategies (eg, anthracycline-free, high-dose cytarabine-based regimens). 
 • In patients with chemotherapy-resistant disease (eg, TP53 mutations, complex cytogenetic aberrations, RUNX1 mutations), alternative therapies instead of anthracycline-containing induction therapy should be considered. 
 • Patients with LVEF decline before and after anthracycline-containing therapy should not be excluded from receiving allogeneic HSCT if the patient is otherwise regarded as fit. Induction therapy in patients with preexisting LVEF decline should be guided by the molecular subtype of AML and can comprise of nonintensive strategies (eg, hypomethylating agent ± venetoclax) depending on the local availability and reimbursement of these therapies. 
Follow-up assessment 
 • Patients who have received anthracycline-containing chemotherapy should have close monitoring of cardiac function, preferably by a cardiologist with experience in managing anthracycline-related left ventricular dysfunction. 
 • Patients should receive longitudinal echocardiograms and clinical reviews for the control of cardiovascular risk factors. 
Integration of cardiology 
 • Comanagement with a cardio-oncologist or a cardiologist with experience in managing anthracycline-related left ventricular dysfunction is strongly encouraged at all stages of therapy and surveillance. 
Considerations for non-anthracycline-based regimens 
 • High probability of induction failure due to AML biology (eg, TP53 mutation, complex cytogenetic aberrations). 
 • Favorable-risk AML and symptomatic heart failure with reduced ejection fraction (LVEF <40%). 
Baseline assessment 
 • Assess for preexisting cardiac comorbidities, risk factors for anthracycline-related left ventricular dysfunction, and frailty before therapy. 
 • Further assessment of clinical signs of HF and cardiac echocardiography should be performed before, during, and after therapy to evaluate for LVEF and if available, global longitudinal strain. 
Prevention 
 • If anthracycline can be administered safely with a prolonged infusion time, this should be considered, especially in patients with preexisting risk factors for anthracycline-related left ventricular dysfunction besides age, or a HF with mid-range reduced ejection fraction. 
Choice of induction regimens according to disease biology and risk factors for anthracycline-related left ventricular dysfunction 
 • In patients with chemotherapy-sensitive leukemia (eg, core-binding factor leukemia, or NPM1-mutated AML), a mild decrease in LVEF should not abrogate a curative approach and standard “7+3” regimen should be considered with the previously mentioned preventive strategy. Other options include reducing daunorubicin dose to 45 mg/m2 or use alternative intensive induction strategies (eg, anthracycline-free, high-dose cytarabine-based regimens). 
 • In patients with chemotherapy-resistant disease (eg, TP53 mutations, complex cytogenetic aberrations, RUNX1 mutations), alternative therapies instead of anthracycline-containing induction therapy should be considered. 
 • Patients with LVEF decline before and after anthracycline-containing therapy should not be excluded from receiving allogeneic HSCT if the patient is otherwise regarded as fit. Induction therapy in patients with preexisting LVEF decline should be guided by the molecular subtype of AML and can comprise of nonintensive strategies (eg, hypomethylating agent ± venetoclax) depending on the local availability and reimbursement of these therapies. 
Follow-up assessment 
 • Patients who have received anthracycline-containing chemotherapy should have close monitoring of cardiac function, preferably by a cardiologist with experience in managing anthracycline-related left ventricular dysfunction. 
 • Patients should receive longitudinal echocardiograms and clinical reviews for the control of cardiovascular risk factors. 
Integration of cardiology 
 • Comanagement with a cardio-oncologist or a cardiologist with experience in managing anthracycline-related left ventricular dysfunction is strongly encouraged at all stages of therapy and surveillance. 
Considerations for non-anthracycline-based regimens 
 • High probability of induction failure due to AML biology (eg, TP53 mutation, complex cytogenetic aberrations). 
 • Favorable-risk AML and symptomatic heart failure with reduced ejection fraction (LVEF <40%). 

The authors gratefully thank Karin Klingel, Department of Cardiopathology, University Hospital Tübingen (Tübingen, Germany), for providing histopathological pictures of anthracycline-related cardiomyopathy.

This work was supported by a research scholarship from the University of Heidelberg (N.R.N.), the Wilmot Research Fellowship Award (K.P.L.), a National Health and Medical Research Council Postgraduate Scholarship (APP10774676) (W.-K.S.), and Eastern Health Clinical School Scholarship (W.-K.S.).

Contribution: N.R.N., K.P.L., A.S.M., K.C., W.-K.S., B.B., and C.O.A. wrote the manuscript; and H.D.K. and W.G.H. supervised the work and edited the manuscript.

Conflict-of-interest disclosure: N.R.N. has received honoraria and travel support from Janssen-Cilag, Medac, and Jazz Pharmaceutical. A.S.M. served on the scientific advisory boards of Agios, Astellas, Abbvie, and Jazz Pharmaceuticals. The remaining authors declare no competing financial interests.

Correspondence: Nina Rosa Neuendorff, Medizinische Klinik V, Universitätsklinik Heidelberg, Im Neuenheimer Feld 410, D-69120 Heidelberg, Germany; e-mail: ninarosa.neuendorff@med.uni-heidelberg.de.

1.
Cancer Statistics Factsheets
SEER
.
Acute Myeloid Leukemia.
National Cancer Institute. Bethesda, MD. http://seer.cancer.gov/statfacts/html/amyl.html. Assessed 17 August
2019
.
2.
Lichtman
MA
.
A historical perspective on the development of the cytarabine (7days) and daunorubicin (3days) treatment regimen for acute myelogenous leukemia: 2013 the 40th anniversary of 7+3
.
Blood Cells Mol Dis
.
2013
;
50
(
2
):
119
-
130
.
3.
Swain
SM
,
Whaley
FS
,
Ewer
MS
.
Congestive heart failure in patients treated with doxorubicin: a retrospective analysis of three trials
.
Cancer
.
2003
;
97
(
11
):
2869
-
2879
.
4.
Von Hoff
DD
,
Layard
MW
,
Basa
P
, et al
.
Risk factors for doxorubicin-induced congestive heart failure
.
Ann Intern Med
.
1979
;
91
(
5
):
710
-
717
.
5.
Dazzi
H
,
Kaufmann
K
,
Follath
F
.
Anthracycline-induced acute cardiotoxicity in adults treated for leukaemia. Analysis of the clinico-pathological aspects of documented acute anthracycline-induced cardiotoxicity in patients treated for acute leukaemia at the University Hospital of Zurich, Switzerland, between 1990 and 1996
.
Ann Oncol
.
2001
;
12
(
7
):
963
-
966
.
6.
Yang
SC
,
Chuang
MH
,
Li
DK
.
The development of congestive heart failure and ventricular tachycardia after first exposure to idarubicin in a patient with acute myeloid leukaemia
.
Br J Clin Pharmacol
.
2010
;
69
(
2
):
209
-
211
.
7.
Hayek
ER
,
Speakman
E
,
Rehmus
E
.
Acute doxorubicin cardiotoxicity
.
N Engl J Med
.
2005
;
352
(
23
):
2456
-
2457
.
8.
Steinberg
JS
,
Cohen
AJ
,
Wasserman
AG
,
Cohen
P
,
Ross
AM
.
Acute arrhythmogenicity of doxorubicin administration
.
Cancer
.
1987
;
60
(
6
):
1213
-
1218
.
9.
Hequet
O
,
Le
QH
,
Moullet
I
, et al
.
Subclinical late cardiomyopathy after doxorubicin therapy for lymphoma in adults
.
J Clin Oncol
.
2004
;
22
(
10
):
1864
-
1871
.
10.
Cardinale
D
,
Colombo
A
,
Bacchiani
G
, et al
.
Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy
.
Circulation
.
2015
;
131
(
22
):
1981
-
1988
.
11.
Alexander
J
,
Dainiak
N
,
Berger
HJ
, et al
.
Serial assessment of doxorubicin cardiotoxicity with quantitative radionuclide angiocardiography
.
N Engl J Med
.
1979
;
300
(
6
):
278
-
283
.
12.
Plana
JC
,
Galderisi
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
(
9
):
911
-
939
.
13.
Boyd
A
,
Stoodley
P
,
Richards
D
, et al
.
Anthracyclines induce early changes in left ventricular systolic and diastolic function: a single centre study
.
PLoS One
.
2017
;
12
(
4
):
e0175544
.
14.
Thavendiranathan
P
,
Poulin
F
,
Lim
KD
,
Plana
JC
,
Woo
A
,
Marwick
TH
.
Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: a systematic review
.
J Am Coll Cardiol
.
2014
;
63
(
25 25 Pt A
):
2751
-
2768
.
15.
Renu
K
,
VG
A
,
Pichiah
T
,
Arunachalam
S
.
Molecular mechanism of doxorubicin-induced cardiomyopathy—an update
.
Eur J Pharmacol
.
2018
;
818
:
241
-
253
.
16.
Tan
TC
,
Neilan
TG
,
Francis
S
,
Plana
JC
,
Scherrer-Crosbie
M
.
Anthracycline-induced cardiomyopathy in adults
.
Compr Physiol
.
2015
;
5
(
3
):
1517
-
1540
.
17.
Zhang
S
,
Liu
X
,
Bawa-Khalfe
T
, et al
.
Identification of the molecular basis of doxorubicin-induced cardiotoxicity
.
Nat Med
.
2012
;
18
(
11
):
1639
-
1642
.
18.
Zamorano
JL
,
Lancellotti
P
,
Rodriguez Muñoz
D
, et al;
ESC Scientific Document Group
.
2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: the Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC)
.
Eur Heart J
.
2016
;
37
(
36
):
2768
-
2801
.
19.
Lubieniecka
JM
,
Graham
J
,
Heffner
D
, et al
.
A discovery study of daunorubicin induced cardiotoxicity in a sample of acute myeloid leukemia patients prioritizes P450 oxidoreductase polymorphisms as a potential risk factor
.
Front Genet
.
2013
;
4
:
231
.
20.
Wouters
KA
,
Kremer
LC
,
Miller
TL
,
Herman
EH
,
Lipshultz
SE
.
Protecting against anthracycline-induced myocardial damage: a review of the most promising strategies
.
Br J Haematol
.
2005
;
131
(
5
):
561
-
578
.
21.
Geisberg
CA
,
Sawyer
DB
.
Mechanisms of anthracycline cardiotoxicity and strategies to decrease cardiac damage
.
Curr Hypertens Rep
.
2010
;
12
(
6
):
404
-
410
.
22.
Skubitz
KM
,
Blaes
AH
,
Konety
SH
,
Francis
GS
.
Cardiac safety profile of patients receiving high cumulative doses of pegylated-liposomal doxorubicin: use of left ventricular ejection fraction is of unproven value
.
Cancer Chemother Pharmacol
.
2017
;
80
(
4
):
787
-
798
.
23.
Lahtinen
R
,
Kuikka
J
,
Nousiainen
T
,
Uusitupa
M
,
Länsimies
E
.
Cardiotoxicity of epirubicin and doxorubicin: a double-blind randomized study
.
Eur J Haematol
.
1991
;
46
(
5
):
301
-
305
.
24.
Feijen
EAM
,
Leisenring
WM
,
Stratton
KL
, et al
.
Derivation of anthracycline and anthraquinone equivalence ratios to doxorubicin for late-onset cardiotoxicity
.
JAMA Oncol
.
2019
;
5
(
6
):
864
-
871
.
25.
Aapro
M
,
Bernard-Marty
C
,
Brain
EG
, et al
.
Anthracycline cardiotoxicity in the elderly cancer patient: a SIOG expert position paper
.
Ann Oncol
.
2011
;
22
(
2
):
257
-
267
.
26.
Negishi
K
,
Negishi
T
,
Haluska
BA
,
Hare
JL
,
Plana
JC
,
Marwick
TH
.
Use of speckle strain to assess left ventricular responses to cardiotoxic chemotherapy and cardioprotection
.
Eur Heart J Cardiovasc Imaging
.
2014
;
15
(
3
):
324
-
331
.
27.
Drafts
BC
,
Twomley
KM
,
D’Agostino
R
Jr.
, et al
.
Low to moderate dose anthracycline-based chemotherapy is associated with early noninvasive imaging evidence of subclinical cardiovascular disease
.
JACC Cardiovasc Imaging
.
2013
;
6
(
8
):
877
-
885
.
28.
Almuwaqqat
Z
,
Gaddh
M
,
Schlafer
D
, et al
Incidence and predictors of left ventricular dysfunction among acute myeloid leukemia patients [abstract]. Blood.
2017
;130(suppl 1). Abstract 5025.
29.
Pasvolsky
O
,
Morelli
O
,
Rozovski
U
, et al
.
Anthracycline-induced cardiotoxicity in acute myeloid leukemia patients who undergo allogeneic hematopoietic stem cell transplantation
.
Clin Lymphoma Myeloma Leuk
.
2019
;
19
(
7
):
e343
-
e348
.
30.
Getz
KD
,
Sung
L
,
Ky
B
, et al
.
Occurrence of treatment-related cardiotoxicity and its impact on outcomes among children treated in the AAML0531 clinical trial: a report from the Children’s Oncology Group
.
J Clin Oncol
.
2019
;
37
(
1
):
12
-
21
.
31.
Bosch
X
,
Rovira
M
,
Sitges
M
, et al
.
Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies)
.
J Am Coll Cardiol
.
2013
;
61
(
23
):
2355
-
2362
.
32.
Le Saux
O
,
Falandry
C
.
Toxicity of cancer therapies in older patients
.
Curr Oncol Rep
.
2018
;
20
(
8
):
64
.
33.
Morley
JE
,
Vellas
B
,
van Kan
GA
, et al
.
Frailty consensus: a call to action
.
J Am Med Dir Assoc
.
2013
;
14
(
6
):
392
-
397
.
34.
Kontny
NE
,
Würthwein
G
,
Joachim
B
, et al
.
Population pharmacokinetics of doxorubicin: establishment of a NONMEM model for adults and children older than 3 years
.
Cancer Chemother Pharmacol
.
2013
;
71
(
3
):
749
-
763
.
35.
Nightingale
G
,
Schwartz
R
,
Kachur
E
, et al
.
Clinical pharmacology of oncology agents in older adults: a comprehensive review of how chronologic and functional age can influence treatment-related effects
.
J Geriatr Oncol
.
2019
;
10
(
1
):
4
-
30
.
36.
Crombag
MR
,
Joerger
M
,
Thürlimann
B
,
Schellens
JH
,
Beijnen
JH
,
Huitema
AD
.
Pharmacokinetics of selected anticancer drugs in elderly cancer patients: focus on breast cancer
.
Cancers (Basel)
.
2016
;
8
(
1
):
E6
.
38.
Li
J
,
Gwilt
PR
.
The effect of age on the early disposition of doxorubicin
.
Cancer Chemother Pharmacol
.
2003
;
51
(
5
):
395
-
402
.
39.
Cusack
BJ
,
Young
SP
,
Vestal
RE
,
Olson
RD
.
Age-related pharmacokinetics of daunorubicin and daunorubicinol following intravenous bolus daunorubicin administration in the rat
.
Cancer Chemother Pharmacol
.
1997
;
39
(
6
):
505
-
512
.
40.
El-Demerdash
E
,
Ali
AA
,
El-Taher
DE
,
Hamada
FM
.
Effect of low-protein diet on anthracycline pharmacokinetics and cardiotoxicity
.
J Pharm Pharmacol
.
2012
;
64
(
3
):
344
-
352
.
41.
Østgård
LS
,
Nørgaard
JM
,
Sengeløv
H
, et al
.
Comorbidity and performance status in acute myeloid leukemia patients: a nation-wide population-based cohort study
.
Leukemia
.
2015
;
29
(
3
):
548
-
555
.
42.
Oran
B
,
Weisdorf
DJ
.
Survival for older patients with acute myeloid leukemia: a population-based study
.
Haematologica
.
2012
;
97
(
12
):
1916
-
1924
.
43.
Giles
FJ
,
Borthakur
G
,
Ravandi
F
, et al
.
The haematopoietic cell transplantation comorbidity index score is predictive of early death and survival in patients over 60 years of age receiving induction therapy for acute myeloid leukaemia
.
Br J Haematol
.
2007
;
136
(
4
):
624
-
627
.
44.
Tawfik
B
,
Pardee
TS
,
Isom
S
, et al
.
Comorbidity, age, and mortality among adults treated intensively for acute myeloid leukemia (AML)
.
J Geriatr Oncol
.
2016
;
7
(
1
):
24
-
31
.
45.
Dhopeshwarkar
N
,
Iqbal
S
,
Wang
X
,
Salas
M
.
A retrospective study of comorbidities and complications in elderly acute myeloid leukemia patients in the United States
.
Clin Lymphoma Myeloma Leuk
.
2019
;
19
(
8
):
e436
-
e456
.
46.
Kuhlman
P
,
Isom
S
,
Pardee
TS
, et al
.
Association between glycemic control, age, and outcomes among intensively treated patients with acute myeloid leukemia
.
Support Care Cancer
.
2019
;
27
(
8
):
2877
-
2884
.
47.
Sorror
ML
,
Maris
MB
,
Storb
R
, et al
.
Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT
.
Blood
.
2005
;
106
(
8
):
2912
-
2919
.
48.
Döhner
H
,
Dolnik
A
,
Tang
L
, et al
.
Cytogenetics and gene mutations influence survival in older patients with acute myeloid leukemia treated with azacitidine or conventional care
.
Leukemia
.
2018
;
32
(
12
):
2546
-
2557
.
49.
Erba
HP
.
Prognostic factors in elderly patients with AML and the implications for treatment
.
Hematology Am Soc Hematol Educ Program
.
2007
;
2007
:
420
-
428
.
50.
Morelli
O
,
Pasvolsky
O
,
Vaturi
M
, et al
Anthracycline cardiotoxicity in patients with acute myeloid leukemia: cardiovascular risk assessment, monitoring and management [abstract]. Eur Heart J.
2017
;38 (suppl 1). Abstract 6160.
51.
Durand
M
,
Lacaria
K
,
Sidsworth
M
,
Davis
MK
,
Sanford
D
.
Management of cardiovascular health in acute leukemia: a national survey
.
Leuk Lymphoma
.
2019
;
60
(
12
):
2982
-
2992
.
52.
Curigliano
G
,
Cardinale
D
,
Suter
T
, et al;
ESMO Guidelines Working Group
.
Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines
.
Ann Oncol
.
2012
;
23
(
Suppl 7
):
vii155
-
vii166
.
53.
Armenian
SH
,
Lacchetti
C
,
Barac
A
, et al
.
Prevention and monitoring of cardiac dysfunction in survivors of adult cancers: American Society of Clinical Oncology Clinical Practice Guideline
.
J Clin Oncol
.
2017
;
35
(
8
):
893
-
911
.
54.
Karanth
NV
,
Roy
A
,
Joseph
M
,
de Pasquale
C
,
Karapetis
C
,
Koczwara
B
.
Utility of prechemotherapy echocardiographical assessment of cardiac abnormalities
.
Support Care Cancer
.
2011
;
19
(
12
):
2021
-
2026
.
55.
Armstrong
GT
,
Plana
JC
,
Zhang
N
, et al
.
Screening adult survivors of childhood cancer for cardiomyopathy: comparison of echocardiography and cardiac magnetic resonance imaging
.
J Clin Oncol
.
2012
;
30
(
23
):
2876
-
2884
.
56.
Jolly
MP
,
Jordan
JH
,
Meléndez
GC
,
McNeal
GR
,
D’Agostino
RB
Jr.
,
Hundley
WG
.
Automated assessments of circumferential strain from cine CMR correlate with LVEF declines in cancer patients early after receipt of cardio-toxic chemotherapy
.
J Cardiovasc Magn Reson
.
2017
;
19
(
1
):
59
.
57.
Charbonnel
C
,
Convers-Domart
R
,
Rigaudeau
S
, et al
.
Assessment of global longitudinal strain at low-dose anthracycline-based chemotherapy, for the prediction of subsequent cardiotoxicity
.
Eur Heart J Cardiovasc Imaging
.
2017
;
18
(
4
):
392
-
401
.
58.
Ali
MT
,
Yucel
E
,
Bouras
S
, et al
.
Myocardial strain is associated with adverse clinical cardiac events in patients treated with anthracyclines
.
J Am Soc Echocardiogr
.
2016
;
29
(
6
):
522
-
527
.
59.
Tan
LL
,
Lyon
AR
.
Role of biomarkers in prediction of cardiotoxicity during cancer treatment
.
Curr Treat Options Cardiovasc Med
.
2018
;
20
(
7
):
55
.
60.
Cardinale
D
,
Sandri
MT
,
Colombo
A
, et al
.
Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy
.
Circulation
.
2004
;
109
(
22
):
2749
-
2754
.
61.
Cardinale
D
,
Sandri
MT
,
Martinoni
A
, et al
.
Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy
.
J Am Coll Cardiol
.
2000
;
36
(
2
):
517
-
522
.
62.
Pastore
F
,
Pastore
A
,
Wittmann
G
,
Hiddemann
W
,
Spiekermann
K
.
The role of therapeutic leukapheresis in hyperleukocytotic AML
.
PLoS One
.
2014
;
9
(
4
):
e95062
.
63.
Lee
YJ
,
Lee
H
,
Park
JS
, et al
.
Cardiac troponin I as a prognostic factor in critically ill pneumonia patients in the absence of acute coronary syndrome
.
J Crit Care
.
2015
;
30
(
2
):
390
-
394
.
64.
Kane
RC
,
McGuinn
WD
Jr.
,
Dagher
R
,
Justice
R
,
Pazdur
R
.
Dexrazoxane (Totect): FDA review and approval for the treatment of accidental extravasation following intravenous anthracycline chemotherapy
.
Oncologist
.
2008
;
13
(
4
):
445
-
450
.
65.
Cvetković
RS
,
Scott
LJ
.
Dexrazoxane: a review of its use for cardioprotection during anthracycline chemotherapy
.
Drugs
.
2005
;
65
(
7
):
1005
-
1024
.
66.
Lemez
P
,
Maresová
J
.
Efficacy of dexrazoxane as a cardioprotective agent in patients receiving mitoxantrone- and daunorubicin-based chemotherapy
.
Semin Oncol
.
1998
;
25
(
4 Suppl 10
):
61
-
65
.
67.
Woodlock
TJ
,
Lifton
R
,
DiSalle
M
.
Coincident acute myelogenous leukemia and ischemic heart disease: use of the cardioprotectant dexrazoxane during induction chemotherapy
.
Am J Hematol
.
1998
;
59
(
3
):
246
-
248
.
68.
Vachhani
P
,
Shin
S
,
Baron
J
, et al
.
Dexrazoxane for cardioprotection in older adults with acute myeloid leukemia
.
Leuk Res Rep
.
2017
;
7
:
36
-
39
.
69.
Reichardt
P
,
Tabone
MD
,
Mora
J
,
Morland
B
,
Jones
RL
.
Risk-benefit of dexrazoxane for preventing anthracycline-related cardiotoxicity: re-evaluating the European labeling
.
Future Oncol
.
2018
;
14
(
25
):
2663
-
2676
.
70.
Volkova
M
,
Russell
R
III
.
Anthracycline cardiotoxicity: prevalence, pathogenesis and treatment
.
Curr Cardiol Rev
.
2011
;
7
(
4
):
214
-
220
.
71.
van Dalen
EC
,
van der Pal
HJ
,
Kremer
LC
.
Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy
.
Cochrane Database Syst Rev
.
2016
;
3
:
CD005008
.
72.
Franco
YL
,
Vaidya
TR
,
Ait-Oudhia
S
.
Anticancer and cardio-protective effects of liposomal doxorubicin in the treatment of breast cancer
.
Breast Cancer (Dove Med Press)
.
2018
;
10
:
131
-
141
.
73.
Rivankar
S
.
An overview of doxorubicin formulations in cancer therapy
.
J Cancer Res Ther
.
2014
;
10
(
4
):
853
-
858
.
74.
Latagliata
R
,
Breccia
M
,
Fazi
P
, et al
.
Liposomal daunorubicin versus standard daunorubicin: long term follow-up of the GIMEMA GSI 103 AMLE randomized trial in patients older than 60 years with acute myelogenous leukaemia
.
Br J Haematol
.
2008
;
143
(
5
):
681
-
689
.
75.
Lancet
JE
,
Uy
GL
,
Cortes
JE
, et al
.
CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia
.
J Clin Oncol
.
2018
;
36
(
26
):
2684
-
2692
.
76.
Mayer
LD
,
Tardi
P
,
Louie
AC
.
CPX-351: a nanoscale liposomal co-formulation of daunorubicin and cytarabine with unique biodistribution and tumor cell uptake properties
.
Int J Nanomedicine
.
2019
;
14
:
3819
-
3830
.
77.
Feldman
EJ
,
Lancet
JE
,
Kolitz
JE
, et al
.
First-in-man study of CPX-351: a liposomal carrier containing cytarabine and daunorubicin in a fixed 5:1 molar ratio for the treatment of relapsed and refractory acute myeloid leukemia
.
J Clin Oncol
.
2011
;
29
(
8
):
979
-
985
.
78.
Peyrade
F
,
Jardin
F
,
Thieblemont
C
, et al;
Groupe d’Etude des Lymphomes de l’Adulte (GELA) investigators
.
Attenuated immunochemotherapy regimen (R-miniCHOP) in elderly patients older than 80 years with diffuse large B-cell lymphoma: a multicentre, single-arm, phase 2 trial
.
Lancet Oncol
.
2011
;
12
(
5
):
460
-
468
.
79.
Burnett
AK
,
Milligan
D
,
Goldstone
A
, et al;
United Kingdom National Cancer Research Institute Haematological Oncology Study Group
.
The impact of dose escalation and resistance modulation in older patients with acute myeloid leukaemia and high risk myelodysplastic syndrome: the results of the LRF AML14 trial
.
Br J Haematol
.
2009
;
145
(
3
):
318
-
332
.
80.
Burnett
AK
,
Russell
NH
,
Hills
RK
, et al;
UK NCRI AML Study Group
.
A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients
.
Blood
.
2015
;
125
(
25
):
3878
-
3885
.
81.
Löwenberg
B
,
Ossenkoppele
GJ
,
van Putten
W
, et al;
Swiss Group for Clinical Cancer Research (SAKK) Collaborative Group
.
High-dose daunorubicin in older patients with acute myeloid leukemia [published correction appears in N Engl J Med. 2010;362(12):1155]
.
N Engl J Med
.
2009
;
361
(
13
):
1235
-
1248
.
82.
Brown
EJ
Jr.
,
Chew
PH
,
MacLean
A
,
Gelperin
K
,
Ilgenfritz
JP
,
Blumenthal
M
;
Fosinopril Heart Failure Study Group
.
Effects of fosinopril on exercise tolerance and clinical deterioration in patients with chronic congestive heart failure not taking digitalis
.
Am J Cardiol
.
1995
;
75
(
8
):
596
-
600
.
83.
The Captopril-Digoxin Multicenter Research Group
.
Comparative effects of therapy with captopril and digoxin in patients with mild to moderate heart failure
.
JAMA
.
1988
;
259
(
4
):
539
-
544
.
84.
CONSENSUS Trial Study Group
.
Effects of enalapril on mortality in severe congestive heart failure. Results of the Cooperative North Scandinavian Enalapril Survival Study (CONSENSUS)
.
N Engl J Med
.
1987
;
316
(
23
):
1429
-
1435
.
85.
Erhardt
L
,
MacLean
A
,
Ilgenfritz
J
,
Gelperin
K
,
Blumenthal
M
;
Fosinopril Efficacy/Safety Trial (FEST) Study Group
.
Fosinopril attenuates clinical deterioration and improves exercise tolerance in patients with heart failure
.
Eur Heart J
.
1995
;
16
(
12
):
1892
-
1899
.
86.
Packer
M
,
Coats
AJ
,
Fowler
MB
, et al;
Carvedilol Prospective Randomized Cumulative Survival Study Group
.
Effect of carvedilol on survival in severe chronic heart failure
.
N Engl J Med
.
2001
;
344
(
22
):
1651
-
1658
.
87.
Sahle
BW
,
Owen
AJ
,
Wing
LMH
,
Beilin
LJ
,
Krum
H
,
Reid
CM
;
Second Australian National Blood Pressure Study Management Committee
.
Long-term survival following the development of heart failure in an elderly hypertensive population
.
Cardiovasc Ther
.
2017
;
35
(
6
):
e12303
.
88.
Yusuf
S
,
Pitt
B
,
Davis
CE
,
Hood
WB
Jr.
,
Cohn
JN
;
SOLVD Investigators
.
Effect of enalapril on mortality and the development of heart failure in asymptomatic patients with reduced left ventricular ejection fractions
.
N Engl J Med
.
1992
;
327
(
10
):
685
-
691
.
89.
Salgado
DR
,
Rocco
JR
,
Silva
E
,
Vincent
JL
.
Modulation of the renin-angiotensin-aldosterone system in sepsis: a new therapeutic approach?
Expert Opin Ther Targets
.
2010
;
14
(
1
):
11
-
20
.
90.
Mawri
S
,
Alsafadi
Y
,
Jain
T
, et al
.
Pre-hospital use of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers is associated with improved outcomes in patients hospitalized with septic shock
.
Integrative Clin Cardiol
.
2017
;
1
(
2
):
006
.
91.
Cardinale
D
,
Colombo
A
,
Sandri
MT
, et al
.
Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition
.
Circulation
.
2006
;
114
(
23
):
2474
-
2481
.
92.
Cardinale
D
,
Ciceri
F
,
Latini
R
, et al;
ICOS-ONE Study Investigators
.
Anthracycline-induced cardiotoxicity: a multicenter randomised trial comparing two strategies for guiding prevention with enalapril: the International CardioOncology Society-one trial
.
Eur J Cancer
.
2018
;
94
:
126
-
137
.
93.
Cardinale
D
,
Colombo
A
,
Lamantia
G
, et al
.
Anthracycline-induced cardiomyopathy: clinical relevance and response to pharmacologic therapy
.
J Am Coll Cardiol
.
2010
;
55
(
3
):
213
-
220
.
94.
Laudisio
A
,
Giovannini
S
,
Finamore
P
, et al
.
Use of ACE-inhibitors and quality of life in an older population
.
J Nutr Health Aging
.
2018
;
22
(
10
):
1162
-
1166
.
95.
Ofran
Y
,
Tallman
MS
,
Rowe
JM
.
How I treat acute myeloid leukemia presenting with preexisting comorbidities
.
Blood
.
2016
;
128
(
4
):
488
-
496
.
96.
Bashey
A
,
Liu
L
,
Ihasz
A
, et al
.
Non-anthracycline based remission induction therapy for newly diagnosed patients with acute myeloid leukemia aged 60 or older
.
Leuk Res
.
2006
;
30
(
4
):
503
-
506
.
97.
Vulaj
V
,
Perissinotti
AJ
,
Uebel
JR
, et al
.
The FOSSIL Study: FLAG or standard 7+3 induction therapy in secondary acute myeloid leukemia
.
Leuk Res
.
2018
;
70
:
91
-
96
.
98.
Brunnberg
U
,
Mohr
M
,
Noppeney
R
, et al
.
Induction therapy of AML with ara-C plus daunorubicin versus ara-C plus gemtuzumab ozogamicin: a randomized phase II trial in elderly patients
.
Ann Oncol
.
2012
;
23
(
4
):
990
-
996
.
99.
Kessler
T
,
Mohr
M
,
Müller-Tidow
C
, et al
.
Amsacrine containing induction therapy in elderly AML patients: comparison to standard induction regimens in a matched-pair analysis
.
Leuk Res
.
2008
;
32
(
3
):
491
-
494
.
100.
Borthakur
G
,
Kantarjian
H
,
Wang
X
, et al
.
Treatment of core-binding-factor in acute myelogenous leukemia with fludarabine, cytarabine, and granulocyte colony-stimulating factor results in improved event-free survival
.
Cancer
.
2008
;
113
(
11
):
3181
-
3185
.
101.
Quintás-Cardama
A
,
Ravandi
F
,
Liu-Dumlao
T
, et al
.
Epigenetic therapy is associated with similar survival compared with intensive chemotherapy in older patients with newly diagnosed acute myeloid leukemia
.
Blood
.
2012
;
120
(
24
):
4840
-
4845
.
102.
Welch
JS
,
Petti
AA
,
Miller
CA
, et al
.
TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes
.
N Engl J Med
.
2016
;
375
(
21
):
2023
-
2036
.
103.
Kuendgen
A
,
Müller-Thomas
C
,
Lauseker
M
, et al
.
Efficacy of azacitidine is independent of molecular and clinical characteristics—an analysis of 128 patients with myelodysplastic syndromes or acute myeloid leukemia and a review of the literature
.
Oncotarget
.
2018
;
9
(
45
):
27882
-
27894
.
104.
Short
NJ
,
Kantarjian
HM
,
Loghavi
S
, et al
.
Treatment with a 5-day versus a 10-day schedule of decitabine in older patients with newly diagnosed acute myeloid leukaemia: a randomised phase 2 trial
.
Lancet Haematol
.
2019
;
6
(
1
):
e29
-
e37
.
105.
US National Library of Medicine
. “Indaction” vs “3+7 induction in AML. https://clinicaltrials.gov/ct2/show/NCT02172872. Accessed 17 August 2019.
106.
DiNardo
CD
,
Pratz
K
,
Pullarkat
V
, et al
.
Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia
.
Blood
.
2019
;
133
(
1
):
7
-
17
.
107.
Wei
A
,
Strickland
SA
,
Hou
J-Z
, et al
.
Venetoclax with low-dose cytarabine induces rapid, deep, and durable responses in previously untreated older adults with AML ineligible for intensive chemotherapy
.
Blood
.
2018
;
132
(
suppl 1
).
Abstract 284
.
108.
Ohanian
M
,
Garcia-Manero
G
,
Levis
M
, et al
.
Sorafenib combined with 5-azacytidine in older patients with untreated FLT3-ITD mutated acute myeloid leukemia
.
Am J Hematol
.
2018
;
93
(
9
):
1136
-
1141
.
109.
Brunner
AM
,
Li
S
,
Fathi
AT
, et al
.
Haematopoietic cell transplantation with and without sorafenib maintenance for patients with FLT3-ITD acute myeloid leukaemia in first complete remission
.
Br J Haematol
.
2016
;
175
(
3
):
496
-
504
.
110.
Uy
GL
,
Mandrekar
SJ
,
Laumann
K
, et al
.
A phase 2 study incorporating sorafenib into the chemotherapy for older adults with FLT3-mutated acute myeloid leukemia: CALGB 11001
.
Blood Adv
.
2017
;
1
(
5
):
331
-
340
.
111.
Serve
H
,
Krug
U
,
Wagner
R
, et al
.
Sorafenib in combination with intensive chemotherapy in elderly patients with acute myeloid leukemia: results from a randomized, placebo-controlled trial
.
J Clin Oncol
.
2013
;
31
(
25
):
3110
-
3118
.
112.
Stein
EM
,
DiNardo
CD
,
Pollyea
DA
, et al
.
Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia
.
Blood
.
2017
;
130
(
6
):
722
-
731
.
113.
DiNardo
CD
,
Stein
EM
,
de Botton
S
, et al
.
Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML
.
N Engl J Med
.
2018
;
378
(
25
):
2386
-
2398
.
114.
Sandhu
KS
,
Aldoss
I
,
Yang
D
, et al
.
A retrospective study of venetoclax-based salvage regimen as a bridge to allogeneic hematopoietic cell transplantation (HCT) in high-risk acute myeloid leukemia (AML) patients
.
Biol Blood Marrow Transplant
.
2019
;
25
(
3
):
S102
-
S103
.
115.
Ossenkoppele
GJ
,
Stussi
G
,
Maertens
J
, et al
.
Addition of bevacizumab to chemotherapy in acute myeloid leukemia at older age: a randomized phase 2 trial of the Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON) and the Swiss Group for Clinical Cancer Research (SAKK)
.
Blood
.
2012
;
120
(
24
):
4706
-
4711
.
116.
Chemnitz
JM
,
Chakupurakal
G
,
Bäßler
M
, et al
.
Pretransplant comorbidities maintain their impact on allogeneic stem cell transplantation outcome 5 years posttransplant: a retrospective study in a single german institution
.
ISRN Hematol
.
2014
;
2014
:
853435
.
117.
Qazilbash
MH
,
Amjad
AI
,
Qureshi
S
, et al
.
Outcome of allogeneic hematopoietic stem cell transplantation in patients with low left ventricular ejection fraction
.
Biol Blood Marrow Transplant
.
2009
;
15
(
10
):
1265
-
1270
.
118.
Fujimaki
K
,
Maruta
A
,
Yoshida
M
, et al
.
Severe cardiac toxicity in hematological stem cell transplantation: predictive value of reduced left ventricular ejection fraction
.
Bone Marrow Transplant
.
2001
;
27
(
3
):
307
-
310
.
119.
Hurley
P
,
Konety
S
,
Cao
Q
,
Weisdorf
D
,
Blaes
A
.
Hematopoietic stem cell transplantation in patients with systolic dysfunction: can it be done?
Biol Blood Marrow Transplant
.
2015
;
21
(
2
):
300
-
304
.
120.
Khouri
MG
,
Klem
I
,
Shenoy
C
,
Sulpher
J
,
Dent
SF
. Screening and monitoring for cardiotoxicity during cancer treatment. In:
Kimmick
GG
,
Lenihan
DJ
,
Sawyer
DB
,
Mayer
EL
,
Hershman
DL
, eds.
Cardio-Oncology: The Clinical Overlap of Cancer and Heart Disease
,
Cham, Switzerland
:
Springer International Publishing
;
2017
:
43
-
80
.
121.
US Food and Drug administration. Drug data base: mitoxantrone. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/019297s030s031lbl.pdf. Accessed 17 August 2019.
122.
Fagard
RH
.
Epidemiology of hypertension in the elderly
.
Am J Geriatr Cardiol
.
2002
;
11
(
1
):
23
-
28
.
123.
Bleumink
GS
,
Knetsch
AM
,
Sturkenboom
MC
, et al
.
Quantifying the heart failure epidemic: prevalence, incidence rate, lifetime risk and prognosis of heart failure The Rotterdam Study
.
Eur Heart J
.
2004
;
25
(
18
):
1614
-
1619
.
124.
Ceia
F
,
Fonseca
C
,
Mota
T
, et al;
EPICA Investigators
.
Prevalence of chronic heart failure in Southwestern Europe: the EPICA study
.
Eur J Heart Fail
.
2002
;
4
(
4
):
531
-
539
.
125.
Ponikowski
P
,
Voors
AA
,
Anker
SD
, et al;
Document Reviewers
.
2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC
.
Eur J Heart Fail
.
2016
;
18
(
8
):
891
-
975
.
126.
Yazdanyar
A
,
Newman
AB
.
The burden of cardiovascular disease in the elderly: morbidity, mortality, and costs
.
Clin Geriatr Med
.
2009
;
25
(
4
):
563
-
577, vii
.
127.
Savji
N
,
Rockman
CB
,
Skolnick
AH
, et al
.
Association between advanced age and vascular disease in different arterial territories: a population database of over 3.6 million subjects
.
J Am Coll Cardiol
.
2013
;
61
(
16
):
1736
-
1743
.
128.
Coresh
J
,
Astor
BC
,
Greene
T
,
Eknoyan
G
,
Levey
AS
.
Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey
.
Am J Kidney Dis
.
2003
;
41
(
1
):
1
-
12
.
129.
Mills
KT
,
Xu
Y
,
Zhang
W
, et al
.
A systematic analysis of worldwide population-based data on the global burden of chronic kidney disease in 2010
.
Kidney Int
.
2015
;
88
(
5
):
950
-
957
.
130.
Vasu
S
,
Kohlschmidt
J
,
Mrózek
K
, et al
.
Ten-year outcome of patients with acute myeloid leukemia not treated with allogeneic transplantation in first complete remission
.
Blood Adv
.
2018
;
2
(
13
):
1645
-
1650
.
131.
Prébet
T
,
Boissel
N
,
Reutenauer
S
, et al;
Core Binding Factor Acute Myeloid Leukemia (CBF AML) intergroup
.
Acute myeloid leukemia with translocation (8;21) or inversion (16) in elderly patients treated with conventional chemotherapy: a collaborative study of the French CBF-AML intergroup
.
J Clin Oncol
.
2009
;
27
(
28
):
4747
-
4753
.
132.
Eisfeld
AK
,
Kohlschmidt
J
,
Mrózek
K
, et al
.
Mutation patterns identify adult patients with de novo acute myeloid leukemia aged 60 years or older who respond favorably to standard chemotherapy: an analysis of Alliance studies
.
Leukemia
.
2018
;
32
(
6
):
1338
-
1348
.
133.
Ostronoff
F
,
Othus
M
,
Lazenby
M
, et al
.
Prognostic significance of NPM1 mutations in the absence of FLT3-internal tandem duplication in older patients with acute myeloid leukemia: a SWOG and UK National Cancer Research Institute/Medical Research Council report
.
J Clin Oncol
.
2015
;
33
(
10
):
1157
-
1164
.
134.
Georgakopoulos
P
,
Roussou
P
,
Matsakas
E
, et al
.
Cardioprotective effect of metoprolol and enalapril in doxorubicin-treated lymphoma patients: a prospective, parallel-group, randomized, controlled study with 36-month follow-up
.
Am J Hematol
.
2010
;
85
(
11
):
894
-
896
.
135.
Kalay
N
,
Basar
E
,
Ozdogru
I
, et al
.
Protective effects of carvedilol against anthracycline-induced cardiomyopathy
.
J Am Coll Cardiol
.
2006
;
48
(
11
):
2258
-
2262
.
136.
Kaya
MG
,
Ozkan
M
,
Gunebakmaz
O
, et al
.
Protective effects of nebivolol against anthracycline-induced cardiomyopathy: a randomized control study
.
Int J Cardiol
.
2013
;
167
(
5
):
2306
-
2310
.
137.
Gulati
G
,
Heck
SL
,
Røsjø
H
, et al
.
Neurohormonal blockade and circulating cardiovascular biomarkers during anthracycline therapy in breast cancer patients: results from the PRADA (Prevention of Cardiac Dysfunction During Adjuvant Breast Cancer Therapy) study
.
J Am Heart Assoc
.
2017
;
6
(
11
):
e006513
.
138.
Silber
JH
,
Cnaan
A
,
Clark
BJ
, et al
.
Enalapril to prevent cardiac function decline in long-term survivors of pediatric cancer exposed to anthracyclines
.
J Clin Oncol
.
2004
;
22
(
5
):
820
-
828
.