Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
Additional content is available online at Elsevier eBooks for Practicing Clinicians
Cardiovascular (CV) disease and cancer are highly prevalent and two major causes of mortality worldwide, resulting in a substantial public health burden. Global estimates suggest 422 million prevalent cases of CV disease. Each year, there are an estimated 17 million incident cases of cancer. CV disease accounts for an estimated 17.9 million deaths and cancer 9.6 million deaths worldwide each year. The overlap in the epidemiology of these two diseases, as well as the shared biologic mechanisms of both CV disease and cancer have led to the birth and maturation of the field of cardio-oncology.
Cardio-oncology is a multidisciplinary field that encompasses the following broad clinical areas: (1) the care of patients with pre-existing CV risk factors or disease who develop cancer; (2) cancer patients and survivors who are at greater propensity for the development of CV risk factors or disease secondary to cancer or cancer therapy; and (3) patients with active or a prior history of cancer who subsequently develop overt CV risk factors or disease. Incident CV disease related to cancer therapy is often termed cardiotoxicity. The diseases encompassed by the term include not only heart failure (HF) (see Chapter 49, Chapter 50, Chapter 51 ), cardiomyopathy (CM), and left ventricular (LV) dysfunction (often referred to as cancer therapeutics related cardiac dysfunction, or CTRCD), but also a broad range of CV disease states, including hypertension (HTN) (see Chapter 26 ), coronary disease and myocardial ischemia (see Chapter 37, Chapter 38, Chapter 40 ), arrhythmia (see Chapter 65, Chapter 66, Chapter 67, Chapter 68 ), pulmonary HTN (see Chapter 88 ), pericardial disease (see Chapter 86 ), valvular disease (see Chapter 72, Chapter 73, Chapter 74, Chapter 75, Chapter 76 ), myocarditis (see Chapter 55 ), peripheral arterial disease (PAD; see Chapter 43 ), and venous and arterial thrombosis.
Both the incidence and significance of cardiotoxicity with cancer therapy are believed to be growing. There are several potential reasons for this. First, survival rates among cancer patients are increasing, potentially related to early detection and more effective treatment regimens, and as a result, the observed “late effects” of cancer therapies are becoming more evident. Second, cancer therapies are rapidly evolving, and while many conventional chemotherapies are still being used, new drug development has led to the development of “targeted” strategies, many of which can also affect fundamental signaling pathways that are necessary for cardiomyocyte and endothelial cell function and homeostasis. This chapter reviews the epidemiology, clinical manifestations, and pathophysiology of CV disease with commonly used chemotherapeutic agents, targeted therapies, immune therapies, hormonal therapy, and radiation therapy (RT) ( Table 56.1 , Fig. 56.1 ). Care of the CV patient prior to, during, and after therapy and strategies to mitigate cardiotoxicity are discussed in Chapter 57 .
Agent | Reported Cardiotoxic Effects | Comments |
---|---|---|
Anthracyclines | ||
Doxorubicin, daunorubicin, epirubicin, idarubicin, mitoxantrone | Cardiac arrhythmias, CM, HF | Risk factors include cumulative dose, although genetic variation may confer increased risk at lower dosages; conventional CV risk factors and disease; age; gender; additional cardiotoxic therapies, including RT or trastuzumab |
Anti-Microtubule Therapies—Taxanes (See Supplement) | ||
Paclitaxel, docetaxel | Arrhythmia, myocardial ischemia | May exacerbate risk of anthracycline cardiotoxicity secondary to pharmacokinetic effects |
Alkylating and Alkylating-Like Agents (See Supplement) | ||
Cyclophosphamide | Myopericarditis, arrhythmias | Rare; CV complications reported primarily at high dosages; emerging data in the transplant setting may suggest increased CV risk. |
Cisplatin, carboplatin, oxaliplatin | Vasculotoxicity, including endothelial dysfunction, arterial vasospasm, HTN | Small studies suggest acute vasculotoxic effects; relationship to long-term events unknown. |
Antimetabolites (See Supplement) | ||
5-Fluorouracil, capecitabine | Coronary vasospasm, myocardial ischemia, infarction, arrhythmias, ECG changes, sudden death | May be related to endothelial injury, vasoconstriction, and vasospasm; typically managed with nitrates and calcium channel blockers |
Monoclonal Antibody Tyrosine Kinase Inhibitors | ||
Bevacizumab | HTN, CM, HF, thrombosis | Low risk of CM or HF |
Trastuzumab | CM, HF | Increased risk of CM and HF with anthracyclines; HTN, obesity, and borderline normal baseline LVEF are also established risk factors; many LVEF declines are reversible, but as noted in clinical trial data, in approximately 20% of patients, reversibility is not seen |
Pertuzumab | CM, HF | Risk of CM and HF remains incompletely defined, but thus far, it has been modest |
Proteasome Inhibitors | ||
Bortezomib and carfilzomib | CM, HF, edema, HTN, acute coronary syndrome, pulmonary hypertension, arrhythmia | Bortezomib is a reversible proteasome inhibitor; carfilzomib is an irreversible proteasome inhibitor; cardiotoxicity rates greater |
Small-Molecule Tyrosine Kinase Inhibitors | ||
Sunitinib | HTN, CM, HF, ischemia, thrombosis | Risk of HTN that tends to occur early; relationship between afterload and CM risk remains to be determined |
Sorafenib | HTN, CM, ischemia, thrombosis | Risk of HTN; also associated with ischemia |
Imatinib | Edema, pericardial effusion | Very low risk of CM |
Nilotinib | Peripheral vascular disease, ischemic heart disease, QT prolongation, cardiometabolic effects | Multi-targeted oral tyrosine kinase inhibitor; cardiometabolic effects include hyperglycemia and hyperlipidemia |
Ponatinib | Peripheral vascular disease, ischemic heart disease, HTN, HF | Multi-targeted oral tyrosine kinase inhibitor |
Dasatinib | Pulmonary HTN, pericardial effusion | Cardiopulmonary status should be evaluated prior to and during therapy |
Ibrutinib | HTN, atrial fibrillation, ventricular arrhythmias, HF, bleeding | Bruton’s tyrosine kinase inhibitor |
Immune-Modulating Agents | ||
Immune checkpoint inhibitors | Myocarditis | Myocarditis is very rare but can be fulminant; worse with combination therapy; typically occurs earlier in course of therapy but can occur at any time |
CAR T cell therapy | Cytokine release syndrome associated adverse CV events (hypotension, arrhythmia) | Treatment for cytokine release syndrome includes tocilizumab |
Androgen-Deprivation Therapy | ||
Leuprolide, goserelin, triptorelin, degarelix, flutamide, bicalutamide | Metabolic syndrome, ischemia, coronary artery disease, HTN | Mixed data regarding an increased risk of adverse CV events with mechanisms of CV disease unclear; patients with pre-existing CV disease may be at a more substantially increased risk |
Estrogen-Receptor Modulators | ||
Tamoxifen | Thrombosis | Favorable effects on lipids |
Aromatase inhibitors (anastrozole, letrozole, exemestane) | Hypercholesterolemia, HTN, HF, combined endpoint of dysrhythmia, valvular disease, and pericarditis | Studies evaluated aromatase inhibitors in comparison to tamoxifen and demonstrated worse CV effects |
Radiation Therapy | Valvular disease, pericardial disease, vascular disease, ischemia, coronary artery disease, CM, HF | Major CV events tend to occur late, although early abnormalities in cardiac function and perfusion are observed; mean heart dose associated with mortality |
The American College of Cardiology and American Heart Association HF Guidelines classify exposure to cardiotoxic therapies, such as anthracyclines, as stage A HF. Reports of the incidence of anthracycline-associated cardiotoxicity have varied widely in the literature, in part, secondary to the variability in the definition of CV outcomes across retrospective analyses and the lack of systematic and rigorously ascertained longitudinal follow-up data, particularly in adults. Historically, cardiotoxicity has been classified as acute, subacute, or chronic although recent studies have substantially challenged this paradigm.
A study of 2625 patients treated with anthracyclines, primarily for breast cancer and hematologic disease, followed over a median of 5.2 years (interquartile range [IQR] 2.6 to 8.0) with serial echocardiography monitoring noted an overall incidence of cardiotoxicity of 9%. Cardiotoxicity was defined as a decrease in the LV ejection fraction (LVEF) of more than 10% from baseline to less than an absolute value of 50%. In 98% of cases, cardiotoxicity was detected within the first year after chemotherapy had been completed, with a median time between the last dose of anthracyclines and the development of cardiotoxicity of 3.5 months (IQR, 3 to 6). In five patients, cardiotoxicity was detected after 5.5 years. The LVEF at the completion of chemotherapy and the cumulative anthracycline dose were independently associated with cardiotoxicity risk. A very small number of patients were hospitalized; the majority were managed as outpatients. HF therapy was initiated in all patients who developed cardiotoxicity, and 82% of the patients recovered their LVEF, either fully or partially. Data from a carefully phenotyped, prospective observational cohort study of breast cancer patients treated with standard dosages of anthracyclines, doxorubicin 240 mg/m 2 , also support the observation that modest declines in quantitative LVEF, on the order of 3% to 4%, occur and are detectable during the 1 to 2 years after the initiation of anthracycline chemotherapy. Altogether, these findings suggest that declines in cardiac function, which may be subclinical, can occur relatively early after chemotherapy completion; these data also challenge the notion of irreversible LVEF declines.
Anthracyclines are a mainstay of therapy in childhood cancers. Cardiotoxicity, when carefully evaluated, is also observed in patients early after exposure to anthracyclines. In clinical trial participants younger than 30 years of age receiving frontline treatment for acute myelogenous leukemia, the cumulative incidence of cardiotoxicity, as defined by National Cancer Institute Common Terminology Criteria for Adverse Events (version 3.0) grade 2 or worse LV systolic dysfunction, was 12%. The standard induction and intensification treatment protocol included daunorubicin and mitoxantrone. In this study, 25% of the cardiotoxicity events were infection associated and 70% of these occurred early, with a median time to cardiotoxicity of 4.3 months (IQR, 3.1 to 5.9) after chemotherapy initiation. Both 5-year event free and overall survival were significantly worsened in patients who suffered from cardiotoxicity. These findings not only indicate an early cardiotoxicity onset, but also suggest treatment-related cardiotoxicity in children results in worse overall outcomes.
Clinical risk factors for anthracycline-induced cardiotoxicity include age of exposure, traditional, modifiable CV risk factors (HTN, diabetes, obesity, and hyperlipidemia), prior chest RT, anthracycline dose, genetic factors, and pre-existing CV disease. In adult survivors of childhood cancer, the prevalence of CM is 4.7% to 10.7%, and increases with age, with an adjusted odds of CM development 2.7 (95% CI 1.1 to 6.9) fold greater compared to patients not exposed to anthracyclines. More recent studies also suggest that the association between anthracycline dose and HF is modified by age of treatment, with a greater relative risk (RR) of CV disease among those who received high-dose anthracycline chemotherapy (≥250 mg/m 2 ) at ≤13 years of age, as compared to children older than 13 years. Here, children that were ≤13 years at diagnosis had a RR of 2.4 to 4.0 fold of any CV disease compared to those older than 13 years. In addition to modifiable CV risk factors and age at anthracycline exposure, concomitant treatment exposures such as RT (>15 Gy) are associated with an increased risk of CV disease. In 1820 adult survivors of childhood cancer treated with anthracyclines, chest-directed RT, or both, there was a high prevalence of subclinical dysfunction in those with an LVEF ≥50%, with abnormal GLS in 28% and diastolic dysfunction in 8.7%, greatest in those who received both anthracyclines and RT.
While multiple studies document a dose-dependent relationship between anthracycline exposure and risk of HF and CM, it is recognized that anthracycline cardiotoxicity can occur at any dose. Older retrospective analyses suggest an incidence of HF, as defined by clinical signs and symptoms, of 1.7% at a cumulative dose of 300 mg/m 2 , 4.7% at 400 mg/m 2 , 15.7% at 500 mg/m 2 , and 48% at 650 mg/m 2 . It is notable that standard errors for many of these estimates are large, given the small sample sizes. Moreover, with respect to dose, emerging data suggest that the equivalency ratios for mitoxantrone relative to doxorubicin is 10.5 (95% CI 6.2 to 19.1), much greater than previously published (0.6 (95% CI 0.4 to 1.0) for daunorubicin, 0.8 (95% CI 0.5 to 2.8) for epirubicin). Data from childhood cancer survivors also indicate that genetic variations in single-nucleotide polymorphisms modify the association between the anthracycline dose and CM risk and confer an increased risk of cardiotoxicity at lower dosages. The genetic determinants of anthracycline cardiotoxicity is an active area of research, with ongoing studies in candidate genes and genome-wide association studies. Additional content on this topic is presented in an online supplement, “Genetics of Anthracycline Cardiotoxicity.”
Several basic mechanisms have been proposed to explain anthracycline-induced cardiotoxicity. The first is formation of reactive oxygen species (ROS) and increased oxidative stress via redox cycling of the quinone moiety of doxorubicin, formation of anthracycline-iron complexes, and topoisomerase-2β (Top2β) inhibition ( Fig. 56.2 ). Furthermore, anthracyclines have been shown to cause impaired calcium signaling and intracellular sequestration affecting myocardial relaxation, a decrease in cardiac progenitor cells, and alterations in neuregulin (NRG)/ErbB signaling. , Recent data also suggest a role for phosphoinositide 3-kinase γ (PI3Kγ) in the pathophysiology of anthracycline cardiotoxicity, perhaps related to the release of mitochondrial DNA by injured organelles and contained autolysosomes. The most widely cited and unifying mechanism is the formation of ROS, leading to oxidative stress and subsequent injury to cardiac myocytes and endothelial cells. , The quinone moiety of the anthracycline enters cells and undergoes redox cycling, generating free radicals via both an enzymatic pathway involving the mitochondrial respiratory chain and also via a nonenzymatic pathway involving direct interactions between anthracyclines and intracellular iron. Toxic hydroxyl radicals from anthracycline-iron complexes act as cytotoxic messengers. This results in impaired mitochondrial function, cellular membrane damage, and cytotoxicity. Nitric oxide synthase (NOS) also contributes to the generation of anthracycline-mediated reactive nitrogen species, worsening nitrosative stress. The formation of ROS may occur via the isozyme Top2, and, more specifically, Top2β, in cardiomyocytes. Mice lacking Top2β are protected from anthracycline-induced DNA damage, cardiomyocyte death, and declines in cardiac function. These findings need to be validated. Interestingly, dexrazoxane, an iron chelator and cardioprotectant, binds to Top2β and results in Top2βdegradation.
Data derived from in vitro and in vivo animal models support the hypothesis that anthracyclines also affect the population of cardiac progenitor cells. Anthracycline chemotherapy may also render cardiomyocytes more susceptible to alterations in NRG-1 and ErbB signaling and downstream prosurvival pathways. , In vitro studies have demonstrated an inhibitory effect of doxorubicin on hypoxia-inducible factor (HIF) and downstream pathways. Anthracyclines also result in impaired diastolic relaxation via calpain-dependent titin proteolysis. More recent studies have used human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) to model the predilection to anthracycline cardiotoxicity in patients, and corroborated decreased cell viability, impaired mitochondrial and metabolic function, impaired calcium handling, decreased antioxidant pathway activity, and increased ROS production as mechanisms of toxicity.
Several other traditional chemotherapeutic agents, including taxanes, alkylating and alkylating-like agents, and antimetabolites can also lead to cardiotoxicity ( see online supplement “Cardiotoxicity of Traditional Chemotherapeutic Agents”).
Understanding the genetic basis of cardiotoxicity is an area of ongoing investigation. There are a number of published studies focused on the use of genome-wide association studies (GWAS), candidate genes, and the use of induced pluripotent stem cells for the study of the inherited basis of disease ( eTables 56.1 and 56.2 ). Processes that have been implicated in the mechanisms of cardiotoxicity and potential causal variants include: (1) the uptake and efflux of anthracyclines by cardiomyocytes (SLC28A3, ATP binding cassette [ABC] transporters); (2) formation of reactive oxygen species (NAD(P)H oxidase, nitric oxide synthase, and cytochrome P450 oxidoreductase [POR]); (3) buffering of reactive oxygen species (catalase, hyaluronan synthase); and (4) anthracycline metabolism (carbonyl reductase (CBR), UDP-glucuronosyltransferase). Interestingly, the risk of cardiotoxicity according to dose may have an inherited basis. In one study, individuals with CBR3:GG genotype were at increased risk of cardiomyopathy (odds ratio 5.48, p = 0.003) at low to moderate, but not high, doses of anthracyclines (<250 mg/m 2 ) compared with patients with CBR3:GA/AA genotypes. Others have identified associations with the CUGBP Elav-like family member 4 (CELF4) genotype and an increased risk of cardiomyopathy at high anthracyclines dosages greater than 300 mg/m 2 . A study of causal genes in dilated cardiomyopathy suggest that titin (TTN) truncating variants may be more prevalent in patients who suffer from cancer cardiomyopathy compared to cancer comparators or healthy controls. GWAS studies have suggested an association between the retinoic acid receptor-gamma (RARG) rs2229774 variant and cardiotoxicity in a European ancestry cohort of childhood cancer patients treated with anthracyclines (P=5.9 × 10 − 8), and confirmed in two additional pediatric cohorts exposed to anthracyclines (P=0.0042 and P=1.2 × 10 − 4). These studies all require validation in larger cohorts of various ancestries, but have been informative in furthering our understanding of disease mechanisms.
Genes Regulating Solute Uptake and Efflux | ||||||
---|---|---|---|---|---|---|
Gene | SNP (Consequence) | Study Reference | Cohorts | Effect of Minor Allele on Cardiotoxicity Risk | P Value | Odds Ratio (95% CI) |
SLC28A3 | rs7853758 (syn) | Visscher 2012 | PC | Decrease | 0.007 (d) | 0.29 (0.11–0.81) |
PC | Decrease | 0.007 (r) | 0.33 (0.13–0.80) | |||
Visscher 2013 | PC | Decrease | 0.058 (r) | 7.98 (1.85–34.4) | ||
Sági 2018 | PC | Decrease | 0.007 (d) | 11.56 (1.98–67.45) | ||
rs885004 (int) | Visscher 2012 | PC | Decrease | 0.017 (d) | 0.33 (0.12–0.91) | |
PC | Decrease | 0.006 (r) | 0.29 (0.11–0.79) | |||
SLC22A17 | rs4982753 (n-c) | Visscher 2015 | PC | Decrease | 0.008 (d) | 0.52 (0.31–0.85) |
PC | Decrease | 0.007 (r) | 0.39 (0.19–0.81) | |||
SLC22A7 | rs4149178 (3’UTR) | Visscher 2015 | PC | Decrease | 0.003 (d) | 0.41 (0.21–0.77) |
PC | Decrease | 0.047 (r) | 0.39 (0.14–1.05) | |||
ABCC1 | rs45511401 (Gly671Val) | Wojnowski 2005 | NHL | Increase | 0.001 | 3.6 (1.6–8.4) |
rs246221 (syn) | Vulsteke 2015 | BC | Increase | 0.02 | 1.6 (1.1–2.3) | |
Semsei 2012 | PC | Decrease | 0.027 | Not specified | ||
rs3743527 (3’UTR) | Semsei 2012 | PC | Increase | 0.001 | Not specified | |
rs4148350 (int) | Visscher 2012 | PC | Increase | 0.00003 | 11.86 (3.37–41.76) | |
ABCC2 | rs8187694 (Val1188Glu) / rs8187710 (Cys1515Tyr) | Wojnowski 2005 | NHL | Increase | 0.049 | 2.3 (1.0–5.4) |
rs8187710 (Cys1515Tyr) | Armenian 2013 | NHL,HL | Increase | 0.02 | 5.22 (1.92–13.84) | |
Aminkeng 2015 | PC | Increase | 0.021 | 4.3 (1.4–13.8) | ||
ABCC5 | rs7627754 (n-c) | Krajinovic 2017 | PC | Increase | <0.0001 | Not specified |
PC | Increase | <0.04 | Not specified | |||
ABCB1 | rs2235047 (int) | Serie 2017 | BC | Increase | 0.018 | Not specified |
Visscher 2012 | PC | Increase | 0.011 | 4.03 (1.32–12.33) | ||
rs1045642 (syn) | Hertz 2016 | BC | Decrease | 0.049 | 0.48 (0.23–1.00) | |
ABCB4 | rs1149222 (int) | Visscher 2012 | PC | Decrease | 0.012 | 2.24 (1.18–4.24) |
Genes Regulating Reactive Oxygen Species (ROS) Production and Buffering | ||||||
---|---|---|---|---|---|---|
Gene | SNP (Consequence) | Study Reference | Cohorts | Effect of Minor Allele on Cardiotoxicity Risk | P Value | Odds Ratio (95% CI) |
RAC2 | rs13058338 (int) | Wojnowski 2005 | NHL | Increase | 0.002 | 2.6 (1.3–5.1) |
Armenian 2013 | NHL,HL | Increase | 0.02 | 2.61 (1.46–4.69) | ||
NCF4 | rs1883112 (n-c) | Wojnowski 2005 | NHL | Increase | 0.007 | 2.5 (1.3–5.0) |
Rossi 2009 | DLBCL | Decrease | 0.023 | 0.37 (0.16–0.87) | ||
CYBA | rs4673 (Tyr72His) | Wojnowski 2005 | NHL | Increase | 0.01 | 1.9 (NA) |
Cascales 2013 | varied | Decrease | 0.039 | 0.11 (0.20–0.63) | ||
NOS3 | rs1799983 (Glu298Asp) | Krajinovic 2017 | PC | Decrease | 0.002 | NA |
CAT | rs10836235 (int) | Rajić 2009 | PC | Decrease | 0.02 | 0.284 (0.093–0.867) |
POR | rs13240755 (int) | Lubieniecka 2013 | PC | Increase | 0.014 | 3.18 (1.223–8.27) |
HAS3 | rs2232228 (syn) | Wang 2014 | PC | Decrease | 0.003 (d) | 8.9 (2.1–37.5) |
Decrease | 0.04 (r) | 4.5 (1.1–18.7) | ||||
Leger 2016 | HCT | Decrease | 0.02 | 21.8 (1.2–386.4) |
Genes Regulating Metabolic Transformations | ||||||
---|---|---|---|---|---|---|
Gene | SNP (Consequence) | Study Reference | Cohorts | Effect of Minor Allele on Cardiotoxicity Risk | P Value | Odds Ratio (95% CI) |
CBR3 | rs1056892 (Val244Met) | Blanco 2012 | PC | Decrease | 0.006 | 3.3 (1.41–7.73) |
Hertz 2016 | BC | Increase | 0.012 | 2.50 (1.22–5.11) | ||
Serie 2017 | BC | Unspecified | 0.004 | 0.84 (0.26–1.41) | ||
CYP3A5 | rs4646450 (int) | Sági 2018 | PC | Increase | 0.004 | 7.25 (1.83–28.78) |
UGT1A6 | rs17863783 (syn) | Visscher 2012 | PC | Increase | 0.04 (c1) | 4.09 (1.03–16.17) |
Increase | 0.075 (c2) | 3.96 (0.92–17.02) | ||||
Increase | 0.0059 (c1,2) | 3.68 (1.45–9.30) | ||||
Visscher 2013 | PC | Increase | 0.0062 (c3) | 7.98 (1.85–34.4) | ||
Increase | 0.0002 (c1,2,3) | 4.30 (1.97–9.36) | ||||
Leger 2016 | HCT | Increase | 0.001 | 19.5 (3.5–110.5) | ||
UGT1A6 | rs6759892 (Ser7Ala) | Visscher 2012 | PC | Increase | 0.0009 (c2) | 2.52 (1.44–4.42) |
Increase | 0.003 (c1,2) | 1.77 (1.20–2.61) | ||||
UGT2B7 | rs7668258 (int) | Li 2019 | BC | Decrease | 0.004 | 0.259 (0.103–0.651) |
GSTP1 | rs1695 (Ile105Val) | Windsor 2012 | PC | Increase | 0.011 | 4.8 (1.4–16.4) |
Study | Design | Patients & major oncologic diagnoses | Case definition or phenotype | Discovery SNPs (P<10 -5 ) | Replicated SNPs and P |
---|---|---|---|---|---|
Aminkeng 2015 | Case-control d = 1, r =1 d: 280 (32 cases, 248 controls). r : 96 (22 cases, 74 controls) | Children ALL,HL/NHL,ES,NB,WT | CTCAEv3-defined cardiac compromise or SF≤24% | rs6895189 (CTNND2, DNAH5), rs7731918 (CTNND2, DNAH5), rs2081944 (CTNND2, DNAH5), rs10085086 (CTNND2, DNAH5), rs15736 (WDR4), rs6586252 (WDR4), rs8133752 (WDR4), rs4381672 (ZNF521), rs4275929 (ZNF521), rs4519409 (ZNF521), rs358224 (GBA3, PPARGC1A), rs412218 (GBA3, PPARGC1A), rs11946006 (GBA3, PPARGC1A), rs7676830 (GBA3, PPARGC1A), rs2282889 (SP4), rs2229774 (RARG), rs9323880 (RIN3), rs7042745 (NCRNA00032) | rs2229774 (RARG); 0.0043 and 1.2 × 10 −4 |
Wang 2016 | Case-control d = 1, r = 1 d : 221 (112 cases, 219 controls). r: 75 (75 cases) | Children HL/NHL, SA, ALL/AML | AHA-defined cardiac compromise or EF≤40% and/or SF≤28% | rs1786814 (CELF4) ∗ | rs1786814 (CELF4); 0.046 |
Schneider 2017 | Case-control d = 1, r =2 d: 3169 (68 cases, 3101 controls). r1: 930 (47 cases, 883 controls). r2: 322 (24 cases, 298 controls) | Adults BC | Cardiologist-adjudicated HF | rs2184559, rs12614202 (LRRTM4), rs12480103, rs9557321 (CLYBL), rs7596623, rs6883259 (ADCY2), rs766643 (ADCY2), rs12545665, rs28714259, rs28684656, rs4058287 (NEDD4L) | rs28714259 (intergenic); 0.04 and 0.018 |
Wells 2017 | Quantitative d = 1, r =1 d: 385. r: 181 | Adults NHL, BC, AML | Maximal change in LVEF | rs10443221 ∗∗ (HOOK1, CYP2J2, C1orf187), rs7542939 (PRDM2), rs78000710 (LRRTM4), rs7635904 (TPRG1), rs990963 (LRRTM4), rs7801891 (AHR, AGR3), rs11066861 (RBM19), rs34223702 (LINC00379), rs7179489 (FAM189A1), rs55669225 (LINC00917), rs12984615 (URI1) | rs7542939 (PRDM2); 0.01 and 6.5 × 10 −7 by meta-analysis |
Serie 2017 | Quantitative d = 1, r =1 d: 800, r : 391 | Adults BC | Maximal change in LVEF | LDB2, BRINP1, RAB22A, TRPC6, LINC01060, intergenic region on chromosome 6 |
The taxanes, paclitaxel (Taxol) and its semisynthetic analogue docetaxel (Taxotere), disrupt microtubular networks as their mechanism of antitumor activity. Used alone, these drugs have relatively little cardiotoxicity; there may be predominantly asymptomatic bradycardia and atrioventricular block.
Cyclophosphamide, used in the treatment of breast cancer and hematologic malignancies, is typically well tolerated. At higher dosages, greater than 100 mg/kg, there have been case reports of hemorrhagic myocarditis, tachyarrhythmia, HF, and pericardial disease. Emerging data on the use of cyclophosphamide in the post-transplant setting may suggest increased CV risk.
Platinum-based agents, often considered alkylating-like agents, are commonly used in germ-cell testicular cancer, as well as ovarian, lung, and breast cancers and other solid tumors. Primary CV concerns with platinum-based therapies have been related to vasculotoxicity, which perhaps has been most well studied in the testicular cancer population. Cases of acute arterial occlusive events, including myocardial infarction and stroke, have been reported in patients receiving cisplatin. In retrospective analyses, the risk of thromboembolic events is elevated within the first year after cancer diagnosis. There is also a hypothesized late risk of cardiotoxicity. One epidemiologic study in testicular cancer survivors with a median observation time of 19 years reported a 5.7-fold increased risk of coronary artery disease and 2.3-fold increased risk of atherosclerotic disease (coronary, cerebrovascular, and peripheral arterial disease) in patients receiving platinum-based chemotherapy regimens compared with patients receiving no chemotherapy. These effects were worsened in patients who received concomitant RT. Platinum results in direct endothelial damage and increased platelet reactivity, which may potentially explain these clinical observations. Platinum levels also remain detectable in the plasma of patients up to 20 years after therapeutic exposure, raising a question as to whether or not prolonged exposure worsens toxicity, although this remains unknown.
5-Fluorouracil (5FU) and its oral analog, capecitabine (Xeloda), are used in the treatment of many solid tumors, including gastrointestinal, breast, head and neck, and pancreatic cancers. CV effects include myocardial ischemia potentially related to vasospasm, as well as cardiac arrhythmias, HTN, hypotension, HF, CM, and even cases of cardiac arrest. A review of 12 Phase II and III clinical trials of 5FU and capecitabine reported the following CV adverse events across these studies: dyspnea in up to 16% of the participants; arrhythmias in less than 1% to 6%; and angina, ischemia, and elevated troponin in less than 1% to 5%. In vitro and in vivo studies suggest that 5FU is associated with endothelial injury, vasospasm and vasoconstriction, and interstitial fibrosis. Capecitabine (Xeloda) is associated with a 6.5% incidence of cardiac events, defined by angina in 4.6%, myocardial infarction, ventricular tachycardia, and sudden death. One retrospective analysis from the SEER Medicare database of individuals greater than 65 years of age with colon cancer suggested that treatment with capecitabine was associated with an increased risk of HF compared to 5FU. Conversely, 5FU treatment was associated with a greater risk of stroke or myocardial infarction compared to capecitabine. Moreover, there was a significant interaction between HTN and chemotherapy and the development of CVD. In a prospective study of 106 patients treated with 5FU, 9% (8.5%) had symptoms of cardiotoxicity, presenting primarily as angina and electrocardiographic changes, as well as elevations in N-terminal pro hormone natriuretic peptide (NT-proBNP).
Proteasome inhibitors, including bortezomib and carfilzomib, are used in the treatment of relapsed or refractory and newly diagnosed cases of multiple myeloma. Bortezomib is a reversible nonselective inhibitor of proteasomes that blocks chymotrypsin-like activity at the 26S proteasome; carfilzomib is an irreversible selective inhibitor of proteasomes that blocks chymotrypsin activity of the 20S proteasome.
The reported incidence of all-grade and grade 3 and higher adverse CV events with these proteasome inhibitors from a meta-analysis of Phase 1 to 3 clinical trials was 18.1% and 8.2%, respectively. HF (4.1%) and HTN (12.2%) were the most common adverse CV events, followed by arrhythmias (2.4%) and ischemia (1.8%). In a multi-center, longitudinal prospective cohort study of 95 patients, the rates of adverse CV events were much greater, occurring in 50.7% in patients receiving carfilzomib and 16.7% receiving bortezomib. Most occurred during carfilzomib therapy; HF and HTN were similarly noted as most common, with grade 3 and 4 HF and HTN events occurring in 20% and 23%, respectively. Arrhythmia, acute coronary syndrome, and pulmonary HTN were also noted. In case series, both HF with preserved and reduced ejection fraction have been reported. Predictors of adverse CV events with carfilzomib include a history of HF, baseline diastolic dysfunction, higher doses of carfilzomib, and abnormal NT-proBNP levels at baseline or during therapy. It is postulated that proteasome inhibitors alter protein homeostasis and reduce phosphorylation of AMPKalpha and downstream autophagy related proteins, such as Raptor.
Immune checkpoint inhibitors have become the standard of care in the treatment of a variety of solid and liquid tumors, revolutionizing cancer care, replacing cytotoxic therapies for many malignancies. These monoclonal antibodies, which target cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and the programmed death receptor (PD-1) and its ligand PD-L1, are associated with a risk of immune-related adverse events. In a retrospective analysis of 448 patients with advanced melanoma treated in Phase I-III trials, 94.9% of treated patients experienced adverse events over a median follow-up of 13.2 months, with 55.4% being grade 3 to 4. Dermatologic, gastrointestinal, endocrine, hepatic, and pulmonary adverse events are most common. Myocarditis, however, occurs much less frequently, on the order of 0.06% to 1%, , but is notable given its potential morbidity and mortality.
The data related to immunotherapy cardiotoxicity are still emerging. Many published studies are derived from retrospective cohort studies or case series. These suggest that myocarditis typically occurs early, at a median time of 34 to 65 days, but over a broad range as there are also published reports of late myocarditis. Additional clinical manifestations include dyspnea, palpitations, and HF. Biomarkers, including troponin and natriuretic peptides have been studied as diagnostic and prognostic tools, as have imaging markers including global longitudinal strain, with associations with adverse cardiac events in small studies.
Combination therapies have been noted to be associated with an approximate twofold increased risk of myocarditis compared to monotherapy. Myositis has also been reported to be common in patients who develop myocarditis. The relevance of other autoimmune diseases or CV disease or risk factors in the development of myocarditis remains incompletely defined. Other toxicities have been reported with immunotherapy, including arrhythmias, pericardial effusions, and CM. Whether these are completely distinct entities from myocarditis is not clear. Moreover, these are emerging data regarding potential associations with accelerated atherosclerosis.
Chimeric antigen receptor (CAR) T cells targeting CD19 are a newer class of therapies that have shown to be highly effective in the treatment of refractory and relapsing hematologic malignancies, including pediatric acute lymphoblastic leukemia and adult large B-cell lymphoma. Toxicities associated with CAR T cell therapy include cytokine release syndrome, a multi-organ system toxicity that occurs with widespread release of inflammatory cytokines and chemokines, including interleukin (IL)-2, soluble IL-2Ra, interferon gamma, IL-6, soluble IL-6R, and granulocyte-macrophage colony-stimulating factor. Adverse CV events most commonly occur with cytokine release syndrome, and include hypotension, arrhythmia, HF, and potentially cardiac death. Treatment for cytokine release syndrome includes tocilizumab.
The treatment of a number of malignant neoplasms has changed dramatically with the advent of targeted therapies. As opposed to traditional chemotherapeutics which target basic cellular processes present in most cells, these therapies target pathways that are dysregulated in cancerous cells. It was hoped that this approach would reduce toxicities typical of conventional chemotherapeutics and be more effective at treating the cancer. However, concerns about cardiotoxicity have surfaced for several agents, likely given the mechanistic commonalities to both CV disease and cancer.
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here