Treatment-related side effects are a major clinical problem in cancer treatment. They lead to reduced compliance to therapy as well as increased morbidity and mortality. Well-known are the sequelae of chemotherapy on the heart, especially in childhood cancer survivors. Therefore, measures to mitigate the adverse events of cancer therapy may improve health and quality of life in patients with cancer, both in the short and long term. The renin–angiotensin system (RAS) affects all hallmarks of cancer, and blockage of the RAS is associated with an improved outcome in several cancer types. There is also increasing evidence that inhibition of the RAS might be able to alleviate or even prevent certain types of cancer treatment–related adverse effects. In this review, we summarize the potential of RAS inhibitors to mitigate cancer treatment–related adverse events, with a special emphasis on chemotherapy-induced cardiotoxicity, radiation injury, and arterial hypertension. Clin Cancer Res; 24(16); 3803–12. ©2018 AACR.

Cancer is a major public health issue, with a projected number of 1,688,780 new cases and 600,920 cancer-related deaths in the United States in 2017 (1). The economic burden of cancer in the European Union rose continuously to €83.2 billion on health care expenditures and €19.1 billion on cancer drugs in 2014, not including production loss due to early death and lost working days (2).

Treatment-related side effects represent a major problem in oncology because they can severely interfere with the patient's quality of life and require dose reductions and treatment delays or even discontinuation of therapy. This reduces the effectiveness of anticancer treatment and ultimately increases morbidity and mortality (3). Side effects also increase the number of emergency room visits and hospitalizations, resulting in increased costs for health care systems and patients. Hence, smart prevention strategies might not only prolong patients' survival and improve their quality of life, but also help to reduce health care costs (4).

The circulating renin–angiotensin system (RAS) plays a pivotal role in maintaining cardiovascular homeostasis as well as fluid and electrolyte balance. Additionally, a local RAS is expressed in many tissues and regulates cellular functions, including growth and metabolism (5, 6). Dysregulation of the local RAS is involved in the pathophysiology of several diseases, such as inflammation and fibrosis (7), and promotes cancer growth and dissemination (8, 9). A recent meta-analysis demonstrated that the intake of angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARB) is significantly associated with improved cancer progression-free and overall survival (10).

Notably, angiotensin II (AngII) is also involved in the development of several cancer treatment–related side effects, such as cardiotoxicity (11), radiation-induced tissue injury (12), and muscle wasting (13–15). Hence, inhibition of AngII/angiotensin II receptor type 1 (AT1R) signaling by renin–angiotensin inhibitors (RASi; i.e., direct renin inhibitors, ACEi, and ARBs) may not only improve the outcome of cancer treatment but also help to treat or prevent certain adverse events. Notably, in addition to inhibiting AngII production, ACEi can also increase angiotensin (1–7) [Ang(1–7)] by blocking its breakdown through ACE. Ang(1–7) is known to counteract many effects of AngII/AT1R signaling (5, 16). ARBs increase AngII levels by blocking the AT1R and thereby also contribute to Ang(1–7) generation from AngII via ACE2 (17, 18). Thus, the beneficial effects of ACEi and ARBs may not only result from inhibiting AngII/AT1R signaling but also partly be mediated by Ang(1–7) (17).

We have recently proposed that RASi-mediated improvement of drug delivery may allow for dose reductions of anticancer drugs without decreasing the therapeutic benefit, which could eventually result in a decreased number of side effects (9). In this review, we discuss the potential of RASi to prevent or improve tumor cachexia as well as cancer treatment–induced adverse events, such as cardiotoxicity, radiation injury, and arterial hypertension.

Several anticancer agents, of which anthracyclines and trastuzumab are widely prescribed, can cause severe and even fatal cardiac side effects, with heart failure due to left ventricular dysfunction (LVD) being the most relevant (19). Of note, the presence of cardiotoxicity not only affects immediate and long-term cardiac outcomes but also limits the therapeutic options in case of disease recurrence. The term “cardiotoxicity” encompasses all side effects affecting the heart, which span the entire cardiac domain, including detectable biomarkers, arrhythmia, structural changes, or clinical symptomatic heart disease. Currently, a general standard definition of cardiotoxicity is lacking, and definitions apply to the specific domain affected [e.g., left ventricular ejection fraction (LVEF) in heart failure; ref. 11].

Cardiotoxicity following chemotherapy or targeted therapy can be divided into acute cardiotoxicity (immediately after administration), early-onset cardiotoxicity (within the first year of treatment), and late-onset cardiotoxicity (several years after chemotherapy; ref. 20). The distinction between early and late onset, however, is more or less artificial, as cardiotoxicity is rather a continuum where some injury occurs/manifests early and others not until later (19).

Late-onset cardiotoxicity, usually preceded by an early asymptomatic period, is of significant importance for pediatric cancer survivors (21). The incidence of anthracycline- and trastuzumab-induced overt heart failure depends on, among other factors, the cumulative dose, concomitant anticancer therapy, and preexisting cardiovascular disease (11, 20). For example, the overall incidence of echocardiographic LVD was 9% after a median period of 5.2 years or 1% to 3% for anti–HER2-targeted therapy (22, 23). Though incidences were much higher when anthracyclines and trastuzumab were concomitantly given, reaching up to 20% after 5 years of follow-up (24).

The generation of reactive oxygen species (ROS)/oxidative stress represents a commonly recognized pathologic mechanism and is a key player in anthracycline-mediated cardiotoxicity (Fig. 1). On the one hand, oxidative radicals are the consequence of the anthracyclines or anthracycline–iron complexes. On the other hand, more recent evidence suggests that topoisomerase-2β in cardiomyocytes leads to increased oxidative stress via its effects on oxidative metabolism and mitochondrial functioning (11, 19). Anthracyclines also appear to affect the neuregulin (NRG)–ErbB (better known as HER) receptor signaling, which is also targeted by trastuzumab. Acute exposure increases the expression of HER2, whereas chronic exposure inhibits HER4 expression, hence disrupting its normal signaling (11). The downstream cellular pathways of the HER2/HER4 heterodimer are responsible for cell performance and survival (25, 26). This also explains why the cardiotoxicity of concomitant treatment with anthracyclines and trastuzumab is increased, because trastuzumab hampers the acute compensatory stress response (26).

Figure 1.

Mechanisms of chemotherapy-induced cardiotoxicity. Anthracyclines induce cellular damage via the formation of ROS and via topoisomerase-2β. Anthracyclines may also affect HER2/HER4 signaling, where HER2 is induced after acute administration, but HER4 is reduced after chronic administration. Trastuzumab inhibits HER2 directly, preventing the formation of the cardioprotective HER2/HER4 heterodimer. AngII is stimulated by trastuzumab and contributes to the formation of ROS. It is also able to inhibit HER2/HER4 signaling via its inhibitory effects on the HER4 ligand neuregulin (NRG). The trophic effects of AngII lead to cardiac remodeling with cellular hypertrophy and fibrogenesis. This is also a consequence of increased arterial blood pressure, which is provoked by inhibition of vascular endothelial growth factor (VEGF). Inhibition of the RAS protects against the negative effects of AngII and stimulates the formation of cardioprotective Ang(1–7).

Figure 1.

Mechanisms of chemotherapy-induced cardiotoxicity. Anthracyclines induce cellular damage via the formation of ROS and via topoisomerase-2β. Anthracyclines may also affect HER2/HER4 signaling, where HER2 is induced after acute administration, but HER4 is reduced after chronic administration. Trastuzumab inhibits HER2 directly, preventing the formation of the cardioprotective HER2/HER4 heterodimer. AngII is stimulated by trastuzumab and contributes to the formation of ROS. It is also able to inhibit HER2/HER4 signaling via its inhibitory effects on the HER4 ligand neuregulin (NRG). The trophic effects of AngII lead to cardiac remodeling with cellular hypertrophy and fibrogenesis. This is also a consequence of increased arterial blood pressure, which is provoked by inhibition of vascular endothelial growth factor (VEGF). Inhibition of the RAS protects against the negative effects of AngII and stimulates the formation of cardioprotective Ang(1–7).

Close modal

When trastuzumab is given alone, it does not cause cell death but rather cardiomyocyte dysfunction by disrupting HER2-mediated signaling, resulting in myocyte stunning or cell-cycle hibernation (19, 27). Therefore, even though debated, trastuzumab-related cardiotoxicity is considered to be reversible (28, 29). Besides the direct inhibition of the prosurvival NRG–HER signaling, trastuzumab increases AngII, which is also a potent inhibitor of NRG. Additionally, AngII enhances ROS production via NADPH oxidase (25). In contrast, Ang(1–7) has cardioprotective effects in heart failure and counteracts the AngII/AT1R axis (ref. 30; Fig. 1).

Preclinical studies demonstrate that ACEi, ARBs, and direct renin inhibitors can protect against chemotherapy-induced cardiotoxicity (31–36) However, clinical data using ACEi or ARBs at standard doses for heart failure reported positive as well as negative/inconclusive results for both substance classes in the preventive setting (Table 1; refs. 37–49). A meta-analysis showed that the prophylactic use of ACEi/ARBs is not able to attenuate the short- to medium-term development of LVD (defined by LVEF) or heart failure [odds ratio 0.24 (95% confidence interval, 0.03–1.73; P = 0.16; 50]. However, results tend to favor ACEi/ARBs, and the lack of statistical significance is likely due to small sample sizes with relative short follow-up and low rate of heart failure (3.6% of all patients; ref. 50).

Table 1.

Randomized controlled trials of RASi to protect against chemotherapy-induced cardiotoxicity

ReferenceCHT type, FU timeTreatment (number of patients)Main findingsOutcome
Primary preventiona 
 Nakamae, 2005 (37) CHOP, 7 days Valsartan 80 mg/d (20), control (20) Control: increased LVEDD, BNP, ANP, QTc interval, QTc dispersion; valsartan: prevented changes except for ANP elevation Positive 
 Georgakopoulos, 2010 (38) Doxorubicin based, 36 months Enalapril (mean, 11 mg/d) (43), metoprolol (mean, 89 mg/d) (42), control (40) No significant difference between the groups in echocardiographic parameters, and development of early/late cardiotoxicity or HF Negative 
 Cadeddu, 2010 (39) Dessì, 2011 (40) Epirubicin based, 18 months Telmisartan 40 mg/d (25), placebo (24) Diastolic impairment, reduced strain rate, and increased IL6 and ROS in placebo group, but not in telmisartan group; echocardiographic findings persisted till 12 months FU Positive 
 Liu, 2013 (41) Anthracycline based, 126 days Candesartan 2.5 mg/d + carvedilol 10 mg/d (20), placebo (20) Attenuated decrease of LVEF in intervention group; increased LVEDD and LVESD in controls, not in intervention group; attenuated ECG changes and arrhythmias in intervention group; reduced troponin in intervention group Positive 
 Bosch, 2013 (42) Intensive CHT for hematologic malignancies, 6 months Enalapril 20 mg/d + carvedilol 50 mg/d (45), control (45) Decreased LVEF only in control group; reduced incidence of combined event (death, HF, or final LVEF<45%) in intervention group Positive 
 Radulescu, 2013 (43) Epirubicin based, until 12 months after end of CHT Perindopril 10 mg/d (68), control (68) Significantly decreased LVEF only in controls; deterioration of LV diastolic function and prolongation of QTc in both groups Inconclusive 
 Janbabai, 2016 (44) Anthracycline based, 6 months Enalapril 20 mg/d (34), placebo (35) Systolic (including LVEF) and diastolic function impaired only in placebo group; troponin I and CK-MB higher in placebo group Positive 
 Gulati, 2016 (45) Anthracycline based ± trastuzumab and radiation, 10–61 weeks Candesartan 32 mg/d + metoprolol 100 mg/d (30), candesartan 32 mg/d (32), metoprolol 100 mg/d (32), placebo (32) No interaction between candesartan and metoprolol; LVEF decline (primary outcome) was reduced by candesartan (but not by metoprolol) vs. placebo; no effect of candesartan on secondary outcomes (RVEF, LV global longitudinal strain, diastolic function, BNP, and troponin I) Positive 
 Boekhout, 2016 (46) Trastuzumb (after anthracycline-based CHT), 92 weeks Candesartan 32 mg/d (103), placebo (103) No difference in LVEF and cardiac events between groups; NT-proBNP and hs-TNT not affected by candesartan Negative 
 Pituskin, 2017 (47) Trastuzumab based, 52 weeks Perindopril 8 mg/d (33), bisoprolol 10 mg/d (31), placebo (30) Primary outcome LV remodeling (LVEDVi) was not prevented by bisoprolol or perindopril; bisoprolol prevented reduction in LVEF; less trastuzumab interruptions due to LVEF drop in perindopril and bisoprolol; multivariate: perindopril and bisoprolol were independent predictors of maintained LVEF Inconclusive 
Secondary preventionb 
 Silber, 2004 (48) Anthracycline based, 34.6 months (mean) Enalapril 0.15 mg/kg/d (69), placebo (66) Enalapril had no effect on exercise performance (including MCI) but reduced LVESWS Inconclusive 
 Cardinale, 2006 (49) High-dose CHT, 12 months Enalapril 20 mg/d (56), control (58) Reduction in LVEF and increase in end-systolic and diastolic volumes only in controls; incidence of cardiac events higher in controls; troponin I normalized within 3 months in enalapril (↑ troponin I at month 3: 0% vs. 21%) Positive 
ReferenceCHT type, FU timeTreatment (number of patients)Main findingsOutcome
Primary preventiona 
 Nakamae, 2005 (37) CHOP, 7 days Valsartan 80 mg/d (20), control (20) Control: increased LVEDD, BNP, ANP, QTc interval, QTc dispersion; valsartan: prevented changes except for ANP elevation Positive 
 Georgakopoulos, 2010 (38) Doxorubicin based, 36 months Enalapril (mean, 11 mg/d) (43), metoprolol (mean, 89 mg/d) (42), control (40) No significant difference between the groups in echocardiographic parameters, and development of early/late cardiotoxicity or HF Negative 
 Cadeddu, 2010 (39) Dessì, 2011 (40) Epirubicin based, 18 months Telmisartan 40 mg/d (25), placebo (24) Diastolic impairment, reduced strain rate, and increased IL6 and ROS in placebo group, but not in telmisartan group; echocardiographic findings persisted till 12 months FU Positive 
 Liu, 2013 (41) Anthracycline based, 126 days Candesartan 2.5 mg/d + carvedilol 10 mg/d (20), placebo (20) Attenuated decrease of LVEF in intervention group; increased LVEDD and LVESD in controls, not in intervention group; attenuated ECG changes and arrhythmias in intervention group; reduced troponin in intervention group Positive 
 Bosch, 2013 (42) Intensive CHT for hematologic malignancies, 6 months Enalapril 20 mg/d + carvedilol 50 mg/d (45), control (45) Decreased LVEF only in control group; reduced incidence of combined event (death, HF, or final LVEF<45%) in intervention group Positive 
 Radulescu, 2013 (43) Epirubicin based, until 12 months after end of CHT Perindopril 10 mg/d (68), control (68) Significantly decreased LVEF only in controls; deterioration of LV diastolic function and prolongation of QTc in both groups Inconclusive 
 Janbabai, 2016 (44) Anthracycline based, 6 months Enalapril 20 mg/d (34), placebo (35) Systolic (including LVEF) and diastolic function impaired only in placebo group; troponin I and CK-MB higher in placebo group Positive 
 Gulati, 2016 (45) Anthracycline based ± trastuzumab and radiation, 10–61 weeks Candesartan 32 mg/d + metoprolol 100 mg/d (30), candesartan 32 mg/d (32), metoprolol 100 mg/d (32), placebo (32) No interaction between candesartan and metoprolol; LVEF decline (primary outcome) was reduced by candesartan (but not by metoprolol) vs. placebo; no effect of candesartan on secondary outcomes (RVEF, LV global longitudinal strain, diastolic function, BNP, and troponin I) Positive 
 Boekhout, 2016 (46) Trastuzumb (after anthracycline-based CHT), 92 weeks Candesartan 32 mg/d (103), placebo (103) No difference in LVEF and cardiac events between groups; NT-proBNP and hs-TNT not affected by candesartan Negative 
 Pituskin, 2017 (47) Trastuzumab based, 52 weeks Perindopril 8 mg/d (33), bisoprolol 10 mg/d (31), placebo (30) Primary outcome LV remodeling (LVEDVi) was not prevented by bisoprolol or perindopril; bisoprolol prevented reduction in LVEF; less trastuzumab interruptions due to LVEF drop in perindopril and bisoprolol; multivariate: perindopril and bisoprolol were independent predictors of maintained LVEF Inconclusive 
Secondary preventionb 
 Silber, 2004 (48) Anthracycline based, 34.6 months (mean) Enalapril 0.15 mg/kg/d (69), placebo (66) Enalapril had no effect on exercise performance (including MCI) but reduced LVESWS Inconclusive 
 Cardinale, 2006 (49) High-dose CHT, 12 months Enalapril 20 mg/d (56), control (58) Reduction in LVEF and increase in end-systolic and diastolic volumes only in controls; incidence of cardiac events higher in controls; troponin I normalized within 3 months in enalapril (↑ troponin I at month 3: 0% vs. 21%) Positive 

Abbreviations: ANP, atrial natriuretic peptide; BNP, brain natriuretic peptide; CHOP, cyclophosphamide/doxorubicin/vincristine/prednisolone; CHT, chemotherapy; CK-MB, creatine kinase-muscle/brain; d, day; ECG, electrocardiogram; FU, follow-up; HF, heart failure; hs-TNT, high-sensitivity troponin T; LV, left ventricular; LVEDD, left-ventricular end-diastolic diameter; LVEDVi, indexed left-ventricular end-diastolic volume; LVESD, left-ventricular end-systolic diameter; LVESWS, left-ventricular end-systolic wall stress; MCI, maximal cardiac index; NT-proBNP, N-terminal of prohormone BNP; RVEF, right-ventricular ejection fraction.

aProphylactic setting.

bAfter detection of subclinical cardiotoxicity (e.g., ↑ troponin I, echocardiographic or ECG abnormalities).

Both ACEi and ARBs are cornerstones in the treatment of heart failure due to their capability to prevent cardiac remodeling and to reduce fibrogenesis (51, 52). Hence, early treatment as soon as (subclinical) cardiotoxicity is detected (via echocardiography or biomarkers) is reasonable. Additionally, almost all cases of anthracycline-induced decline in LVEF are seen in the first 12 months following chemotherapy, implying that cardiotoxicity is a rather early phenomenon (22). Indeed, the sooner the ACEi enalapril alone or in combination with the β-blocker carvedilol was given after detection of LVEF decline following chemotherapy, the larger the potential of full recovery of cardiac function. When initiated after 6 months, recovery was incomplete (53). Real-life data in older patients with follow-up to 5 years confirm the beneficial effects of ACEi and β-blockers, where prompt start (i.e., within 6 months) was associated with lower cardiotoxicity risk and better cardiovascular outcomes (54).

The clinical guidelines of both the American Society of Clinical Oncology and the European Society of Cardiology recommend baseline and periodically surveillance of cardiac dysfunction. Screening includes cardiac biomarkers (troponins and natriuretic peptides) and cardiac imaging (echocardiography and cardiac magnetic resonance). It should be performed in case of clinical suspicion and in those patients at increased risk of developing cardiac dysfunction (20, 55). Both organizations acknowledge the potential or RASi as part of a preventive strategy but refrain from making a recommendation hereupon currently.

A proposed algorithm for anthracycline-related cardiotoxicity suggests enalapril in case of troponin I abnormalities and combined use of ACEi and β-blockers in case of LVD on echocardiography (19, 56). However, this algorithm has not been validated. In general, treatment of chemotherapy-induced LVD and heart failure is similar to conventional heart failure therapy (52) and aims to reduce pathologic left ventricular (LV) remodeling (57, 58). ACEi and ARBs represent a mainstay because they can improve remodeling by reducing LV afterload and by directly antagonizing AngII/angiotensin II receptor type 1 (AT1R)–mediated hypertrophy and fibrosis (58, 59).

Taken together, ACEi/ARBs do have a clear potential to alleviate cardiotoxicity when given prophylactically or as soon as subclinical cardiotoxicity is detected. However, we are awaiting further larger studies. Little is known about the protective effects of long-term RASi use on cardiac outcome after chemotherapy (e.g., in childhood cancer survivors). Table 2 gives an overview of currently ongoing clinical trials.

Table 2.

Ongoing prospective trials of RASi to protect against cancer treatment–related side effects

ClinicalTrials.gov identifier (study acronym)Condition, treatmentDesignInterventionPrimary endpointStatus
Cardiotoxicity 
NCT03127631 (RADICALPC) Prostate cancer, androgen deprivation therapy RCT SOC Composite of death, MI, stroke, HF, or arterial revascularization Recruiting 
   Behavioral interventions (nutrition, exercise, smoking cessation) + ASA + statin + ACEi   
NCT02907021 (SCHOLAR) HER2-positive breast cancer (nonmetastatic) with moderate LV dysfunction, trastuzumab Single arm, phase I ACEi + BB Development of cardiac dose-limiting toxicity Not yet recruiting 
NCT02236806 (SAFE) Breast cancer (nonmetastatic), anthracyclines ± anti-HER2 RCT, phase III Placebo LVEF Unknown 
   Bisoprolol   
   Ramipril   
   Bisoprolol + ramipril   
NCT01904903 (SAFE-HEaRT) HER2-positive breast cancer with mild LV dysfunction, anti-HER2 Single arm, phase II ACEi + BB Completion of therapy without cardiac events or worsening of cardiac function Recruiting 
NCT03265574 (PROACT) Breast cancer, adjuvant epirubicin RCT, phase III SOC Cardiac troponin T release, cardiac function Recruiting 
   Enalapril   
NCT01968200 (ICOS-ONE) Cancer, treatment with anthracyclines RCT, phase III Enalapril (concomitant) Occurrence of cTn elevation Active, not recruiting 
   Enalapril (after biochemical proven injury)   
NCT01009918 HER2-positive breast cancer, trastuzumab RCT, phase III Placebo LVEF Active, not recruiting 
   Carvedilol (extended release)   
   Lisinopril   
Radiotherapy-related toxicities 
NCT01805453 (ASTER) Newly diagnosed glioblastoma, radiotherapy + temozolomide RCT, phase II Placebo Steroid dose needed to control brain edema after radiotherapy Active, not recruiting 
   Losartan   
NCT01754909 Lung cancer, radiotherapy RCT, phase II Placebo Radiation pneumonitis Recruiting 
   Enalapril   
NCT01880528 Lung cancer, radiotherapy RCT Placebo Primary: adverse events of lisinopril Active, not recruiting 
    Secondary: dyspnea, symptoms, QoL  
   Lisinopril   
NCT00004230 Bone marrow or stem cell transplantation, following chemotherapy and radiotherapy RCT, phase III SOC Lung injury Completed 
   Captopril   
Others 
NCT02651415 (PARICCA) Metastatic colorectal cancer, regorafenib Single arm, phase II Perindopril HFSR, AHT Active, not recruiting 
ClinicalTrials.gov identifier (study acronym)Condition, treatmentDesignInterventionPrimary endpointStatus
Cardiotoxicity 
NCT03127631 (RADICALPC) Prostate cancer, androgen deprivation therapy RCT SOC Composite of death, MI, stroke, HF, or arterial revascularization Recruiting 
   Behavioral interventions (nutrition, exercise, smoking cessation) + ASA + statin + ACEi   
NCT02907021 (SCHOLAR) HER2-positive breast cancer (nonmetastatic) with moderate LV dysfunction, trastuzumab Single arm, phase I ACEi + BB Development of cardiac dose-limiting toxicity Not yet recruiting 
NCT02236806 (SAFE) Breast cancer (nonmetastatic), anthracyclines ± anti-HER2 RCT, phase III Placebo LVEF Unknown 
   Bisoprolol   
   Ramipril   
   Bisoprolol + ramipril   
NCT01904903 (SAFE-HEaRT) HER2-positive breast cancer with mild LV dysfunction, anti-HER2 Single arm, phase II ACEi + BB Completion of therapy without cardiac events or worsening of cardiac function Recruiting 
NCT03265574 (PROACT) Breast cancer, adjuvant epirubicin RCT, phase III SOC Cardiac troponin T release, cardiac function Recruiting 
   Enalapril   
NCT01968200 (ICOS-ONE) Cancer, treatment with anthracyclines RCT, phase III Enalapril (concomitant) Occurrence of cTn elevation Active, not recruiting 
   Enalapril (after biochemical proven injury)   
NCT01009918 HER2-positive breast cancer, trastuzumab RCT, phase III Placebo LVEF Active, not recruiting 
   Carvedilol (extended release)   
   Lisinopril   
Radiotherapy-related toxicities 
NCT01805453 (ASTER) Newly diagnosed glioblastoma, radiotherapy + temozolomide RCT, phase II Placebo Steroid dose needed to control brain edema after radiotherapy Active, not recruiting 
   Losartan   
NCT01754909 Lung cancer, radiotherapy RCT, phase II Placebo Radiation pneumonitis Recruiting 
   Enalapril   
NCT01880528 Lung cancer, radiotherapy RCT Placebo Primary: adverse events of lisinopril Active, not recruiting 
    Secondary: dyspnea, symptoms, QoL  
   Lisinopril   
NCT00004230 Bone marrow or stem cell transplantation, following chemotherapy and radiotherapy RCT, phase III SOC Lung injury Completed 
   Captopril   
Others 
NCT02651415 (PARICCA) Metastatic colorectal cancer, regorafenib Single arm, phase II Perindopril HFSR, AHT Active, not recruiting 

Abbreviations: AHT, arterial hypertension; ASA, acetyl salicylic acid; BB, β-blocker; cTn, cardiac troponins; HF, heart failure; HFSR, hand–foot skin reaction; LV, left ventricular; MI, myocardial infarction; QoL, quality of life; RCT, randomized controlled trial; SOC, standard of care.

Arterial hypertension represents a typical and mechanism-dependent “on-target” side effect of antivascular endothelial growth factor (VEGF) therapy and has also been proposed as a biomarker for treatment response (60–62). The induced afterload following arterial hypertension is a risk factor for the development of congestive heart failure, with a 2.69-fold increased risk for patients receiving VEGF receptor tyrosine kinase inhibitors (63). Thus, treatment should start early and resolutely (20).

Several potential pathophysiologic mechanisms have been proposed, including capillary rarefication, downregulation of vasodilators (e.g., nitric oxide, prostacyclin), shift in the pressure–natriuresis curve, and upregulation of vasoconstrictors (e.g., endothelin-1, thromboxanes; refs. 62, 64). The renin–angiotensin–aldosterone system seems not to play a significant role in the pathophysiology of anti–VEGF-mediated hypertension (62), but renin concentration and activity might even be suppressed in patients with anti–VEGF-induced hypertension (65). However, if antihypertensive medication is required, ACEi represent a preferred first-line option, especially in case of concomitant anti–VEGF-induced proteinuria (20, 60, 64, 66). Moreover, treatment of VEGF-induced hypertension with RASi was associated with improved overall survival in metastatic renal cell carcinoma (67).

Notably, based on a small series, others hypothesized that ACEi might reduce the efficacy of bevacizumab by counteracting its antiangiogenic effect (68). This is not in line with other studies suggesting additive effects of RAS inhibition and anti–VEGF-based therapies (9, 69). Unfortunately, a phase II study (NCT01705392) designed to investigate the ability of enalapril to prevent bevacizumab-induced hypertension compared with propranolol was suspended due to insufficient financial support. Hence, even though ACEi represent a first-line option, prospective trials are needed to determine which class of antihypertensive agents is most effective in preventing and treating anti–VEGF-related hypertension.

Pathophysiologically, radiation can induce tissue injury through direct (radiation–DNA interaction) and indirect (production of ROS) DNA damage, and, if not repaired, eventually results in cell injury or death (70). There is some evidence that irradiation upregulates AngII expression in a dose-dependent manner (71, 72). The AngII/AT1R axis contributes to tissue damage and remodeling by upregulation of profibrogenic (e.g., TGFβ) and proinflammatory pathways and by production of ROS via activation of NADPH oxidase (12). Hence, inhibition of AngII/AT1R signaling represents an attractive target to prevent or reduce radiation-induced injury (Fig. 2).

Figure 2.

Mechanisms of radiation-induced tissue injury. Radiation damages cellular DNA, both directly as well as indirectly via the formation of ROS, that eventually results in cell death. This is augmented by the capability of radiation to upregulate AngII that contributes to the formation of ROS. The direct effects of radiation lead to capillary hyperpermeability that can lead to edema/erythema. This can be further augmented by AngII, which increases vascular permeability via the stimulation of VEGF. Increased AngII itself stimulates inflammation and fibrosis of tissues. Inhibition of the RAS can reduce edema and lessens the effects on tissues. Moreover, induction of Ang(1–7) can counteract the effects of AngII.

Figure 2.

Mechanisms of radiation-induced tissue injury. Radiation damages cellular DNA, both directly as well as indirectly via the formation of ROS, that eventually results in cell death. This is augmented by the capability of radiation to upregulate AngII that contributes to the formation of ROS. The direct effects of radiation lead to capillary hyperpermeability that can lead to edema/erythema. This can be further augmented by AngII, which increases vascular permeability via the stimulation of VEGF. Increased AngII itself stimulates inflammation and fibrosis of tissues. Inhibition of the RAS can reduce edema and lessens the effects on tissues. Moreover, induction of Ang(1–7) can counteract the effects of AngII.

Close modal

Indeed, treatment with ACEi or ARBs has demonstrated some efficacy in preventing radiation-induced damage of normal tissue in different organs in preclinical studies (73–76) without compromising tumor response to radiation (77).

ACEi and ARBs might inhibit radiation-induced toxicity not only by their effects on AngII and AT1R but also by increasing Ang(1–7) (16, 17), which indeed seems to have radioprotective effects (78, 79). Noteworthy, both AngII and Ang(1–7) improved hematopoietic recovery after total body irradiation in mice (80).

Data from clinical trials are scarce and inconsistent. Several retrospective studies reported a decreased risk of radiation pneumonitis in lung cancer patients using ACEi/ARBs (81–86), whereas others found no association (87, 88). Interestingly, in one study, a beneficial effect was only seen with ACEi and not with ARB (81).

As these retrospective studies only provide pooled analysis of different ACEi/ARB compounds, no conclusions on the effect of each drug within a RASi subclass can be drawn and no specifications on dosing can be made. However, one can assume that standard doses to treat cardiovascular diseases were used. Unfortunately, a large prospective study using captopril in patients with radiation-treated lung cancer was halted due to insufficient accrual (89).

ACEi use was also associated with reduced risk of radiation-induced proctitis in a retrospective study (90). A prospective noninterventional study found that statins alone and ACEi in combination with statins (but not ACEi alone) reduced gastrointestinal toxicity after pelvic radiation (91). In a small randomized controlled trial of 55 subjects undergoing total body irradiation for hematopoietic stem cell transplantation, captopril at a maximum dose of 25 mg three times a day had no significant effect on the risk of chronic renal failure and on overall and pulmonary mortality (92).

Finally, radiation increases the permeability of the capillary wall and leads to endothelial dysfunction (93). The consequential edema is a frequent and threatening side effect when treating brain tumors. Two retrospective studies showed that the use of ACEi/ARBs was associated with reduced steroid doses for vasogenic edema in glioblastoma via blockage of VEGF production that is abundantly present in those tumors (94, 95). The use of ACEi/ARBs is also protective for the adjacent normal brain tissue by preventing radiation necrosis (96), whereas preclinical evidence demonstrates the capability of an ARB to preserve cognition (97).

Taken together, ACEi/ARBs seem to be protective against the effects of radiation injury in several organ sites. However, prospective trials are needed for further validation. Trials currently testing RASi to prevent radiation injury are listed in Table 2.

Cachexia is a multifactorial catabolic condition characterized by substantial weight and muscle loss (with or without fat loss) due to an underlying disease that cannot be completely reversed by nutritional support (98, 99). Cancer cachexia can reach different stages ranging from pre-cachexia to cachexia and refractory cachexia (99) and should be considered as a partially preventable cancer comorbidity (100). Cachexia has a high prevalence in patients with cancer (30%–60%); accounts for up to 20% of deaths; and represents an independent predictor of mortality, treatment response, and quality of life (101, 102). AngII signaling contributes to muscle wasting by different mechanisms such as induction of muscle protein catabolism through upregulation of the ubiquitin–proteasome proteolytic pathway (13), inhibition of protein synthesis in myotubes (15), and induction of apoptosis in myocytes (14). Although the induction of skeletal myocyte apoptosis is mediated via AT1R (103), the effects of AngII on protein degradation in skeletal muscle are likely mediated via the angiotensin II receptor type 2 (AT2R; ref. 13), known to counteract the effects of AngII via AT1R (5). This suggests that ACEi—by inhibiting AngII formation—may have a more potent protective effect in preventing cancer cachexia than ARBs, which only block AT1R.

Interestingly, in an observational study of older hypertensive women, continuous ACEi use significantly slowed decline in muscle strength (104).

Several preclinical studies have demonstrated that ACEi can control cancer cachexia in different rodent models (13, 105, 106), whereas imidapril had no beneficial effect on cardiac wasting in a rat hepatoma cancer cachexia model (107). In a subcutaneous murine colon cancer model, the direct renin inhibitor aliskiren had potent anti-cachexia effects. Aliskiren antagonized several mechanisms of cancer cachexia, namely, RAS activation, inflammation, oxidative stress, and stimulation of the authophagy–lysosome as well as the ubiquitin–proteasome pathway (108).

In a phase III trial conducted by Ark Therapeutics, the ACEi imidapril attenuated weight loss in non–small cell lung cancer (NSCLC) and colorectal cancer, but not in pancreatic cancer; however, the trial missed its primary endpoint because the significance was gone in pooled analysis (http://www.apmhealtheurope.com/print_story.php?numero=L1135).

Many patients with cancer develop chemotherapy-induced peripheral neuropathy, with long-lasting and refractory pain in a substantial percentage of patients. In a retrospective study analyzing the results of a standardized panel testing sensory and pain sensation of the small fibers of the peripheral nerves (i.e., of the hand), ACEi/ARBs were associated with attenuated sensory loss. The authors suggest an AT2R-mediated neuromodulating effect over the small myelinated fibers of the glabrous skin (109).

An ongoing trial (NCT02651415) is currently investigating the potential of the ACEi perindopril to mitigate the occurrence of hand–foot skin reactions—a well-known side effect of tyrosine kinase inhibitors—in patients with colorectal cancer treated with regorafenib (Table 2).

The renin–angiotensin system is, without doubt, closely intertwined with cancer. Available data suggest that concomitant use of RASi with standard cancer therapy may reduce morbidity and mortality of patients with cancer. Moreover, RASi might also alleviate treatment-related side effects of cancer therapies, such as cardiotoxicity and radiation-induced tissue injury. However, knowledge on the role of RASi herein is currently limited to experimental studies, retrospective analyses, or small prospective studies. Consequently, to date, definitive recommendations cannot be made. Additionally, most studies evaluated the role of ACEi and ARBs in this context, and only little data are available for direct renin inhibitors—the third class of RASi.

The main body of research is related to the potential effects of RASi to reduce cardiotoxicity in anthracycline- or trastuzumab-induced cardiotoxicity. There is a trend that RASi at standard doses could be useful in a prophylactic setting, but unequivocal evidence is lacking. The evidence for treating (sub)clinical heart failure with RASi is more substantial. Therefore, prophylactic treatment with RASi should be limited to selected high-risk patients. Once (sub)clinical heart failure is detected, treatment should be started promptly (20). Similarly, arterial hypertension should be treated aggressively and as early as possible in order to prevent the development of congestive heart failure (20).

Within the field of radiation-induced side effects, available data are scarce. Most studies currently investigating RASi in the setting of radiation are focused on the prevention of radiation pneumonitis, as this seems to be a promising approach.

Notably, by modulating the tumor microenvironment and improving drug delivery to tumors, RASi may also allow for dose reductions of anticancer drugs, including immunotherapy, without decreasing their therapeutic benefit, and this could ultimately decrease toxicity (9).

Finally, even though RASi may help to attenuate various side effects of cancer treatment, decisions for using these drugs are to be made on a case-by-case basis. For example, due to their effects on the glomerular filtration rate, RASi actually increase the risk on platinum-based nephrotoxicity (110).

Because RASi are already FDA approved, have a well-known safety profile, and are relatively inexpensive, they are an appealing option to improve cancer-therapy substantially. Their future potential might grow even further in the quickly evolving landscape of cancer therapeutics. However, there is an obvious need for properly powered randomized controlled trials to evaluate the full potential of RASi in preventing or improving certain cancer treatment–related adverse events. Studies with extended follow-up have the potential to alleviate the long-term side effects.

Prospective studies should also investigate if standard doses of RASi are required in the prevention setting or if even reduced doses have the potential to mitigate cancer treatment–induced adverse events.

M. Pinter reports receiving speakers bureau honoraria from Bayer and Bristol-Myers Squibb, and is a consultant/advisory board member for Bayer, Bristol-Myers Squibb, and Eisai. R.K. Jain reports receiving speakers bureau honoraria from Amgen, holds ownership interest (including patents) in Enlight, Ophthotech, SynDevRx, and Xtuit, and is a consultant/advisory board member for Enlight, Merck, Ophthotech, Pfizer, SPARC, SynDevRx, Tekla Healthcare Investors, Tekla Healthcare Opportunities Fund, Tekla Life Sciences Investors, Tekla World Healthcare Fund, and XTuit. No potential conflicts of interest were disclosed by the other author.

M. Pinter is supported by an Erwin-Schroedinger Fellowship by the Austrian Science Fund (J 3747-B28). R.K. Jain is supported by NCI P01-CA080124, NCI R35-CA197743, NCI R01-CA208205, NCI U01-CA224173, and the Bill and Melinda Gates Foundation (OPP1140482).

The authors want to express their gratitude to Drs. Jeffrey Clark and Melin J. Khandekar for their comments on the article.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2017
.
CA Cancer J Clin
2017
;
67
:
7
30
.
2.
Jönsson
B
,
Hofmarcher
T
,
Lindgren
P
,
Wilking
N
. 
The cost and burden of cancer in the European Union 1995–2014
.
Eur J Cancer
2016
;
66
:
162
70
.
3.
Reulen
RC
,
Winter
DL
,
Frobisher
C
,
Lancashire
ER
,
Stiller
CA
,
Jenney
ME
, et al
Long-term cause-specific mortality among survivors of childhood cancer
.
JAMA
2010
;
304
:
172
9
.
4.
Nolan
MT
,
Plana
JC
,
Thavendiranathan
P
,
Shaw
L
,
Si
L
,
Marwick
TH
. 
Cost-effectiveness of strain-targeted cardioprotection for prevention of chemotherapy-induced cardiotoxicity
.
Int J Cardiol
2016
;
212
:
336
45
.
5.
Paul
M
,
Poyan Mehr
A
,
Kreutz
R
. 
Physiology of local renin–angiotensin systems
.
Physiol Rev
2006
;
86
:
747
803
.
6.
Bader
M
. 
Tissue renin–angiotensin–aldosterone systems: targets for pharmacological therapy
.
Annu Rev Pharmacol Toxicol
2010
;
50
:
439
65
.
7.
Passos-Silva
DG
,
Brandan
E
,
Santos
RAS
. 
Angiotensins as therapeutic targets beyond heart disease
.
Trends Pharmacol Sci
2015
;
36
:
310
20
.
8.
George
AJ
,
Thomas
WG
,
Hannan
RD
. 
The renin–angiotensin system and cancer: old dog, new tricks
.
Nat Rev Cancer
2010
;
10
:
745
59
.
9.
Pinter
M
,
Jain
RK
. 
Targeting the renin–angiotensin system to improve cancer treatment: implications for immunotherapy
.
Sci Transl Med
2017
;
9
.
10.
Sun
H
,
Li
T
,
Zhuang
R
,
Cai
W
,
Zheng
Y
. 
Do renin–angiotensin system inhibitors influence the recurrence, metastasis, and survival in cancer patients?: evidence from a meta-analysis including 55 studies
.
Medicine (Baltimore)
2017
;
96
:
e6394
.
11.
McGowan
JV
,
Chung
R
,
Maulik
A
,
Piotrowska
I
,
Walker
JM
,
Yellon
DM
. 
Anthracycline chemotherapy and cardiotoxicity
.
Cardiovasc Drugs Ther
2017
;
31
:
63
75
.
12.
Robbins
ME
,
Diz
DI
. 
Pathogenic role of the renin–angiotensin system in modulating radiation-induced late effects
.
Int J Radiat Oncol Biol Phys
2006
;
64
:
6
12
.
13.
Sanders
PM
,
Russell
ST
,
Tisdale
MJ
. 
Angiotensin II directly induces muscle protein catabolism through the ubiquitin-proteasome proteolytic pathway and may play a role in cancer cachexia
.
Br J Cancer
2005
;
93
:
425
34
.
14.
Burniston
JG
,
Saini
A
,
Tan
L-B
,
Goldspink
DF
. 
Angiotensin II induces apoptosis in vivo in skeletal, as well as cardiac, muscle of the rat
.
Exp Physiol
2005
;
90
:
755
61
.
15.
Russell
ST
,
Sanders
PM
,
Tisdale
MJ
. 
Angiotensin II directly inhibits protein synthesis in murine myotubes
.
Cancer Lett
2006
;
231
:
290
4
.
16.
Tom
B
,
Dendorfer
A
,
Danser
AHJ
. 
Bradykinin, angiotensin-(1-7), and ACE inhibitors: how do they interact?
Int J Biochem Cell Biol
2003
;
35
:
792
801
.
17.
Schindler
C
,
Bramlage
P
,
Kirch
W
,
Ferrario
CM
. 
Role of the vasodilator peptide angiotensin-(1-7) in cardiovascular drug therapy
.
Vasc Health Risk Manag
2007
;
3
:
125
37
.
18.
Arumugam
S
,
Thandavarayan
RA
,
Palaniyandi
SS
,
Giridharan V
V
,
Arozal
W
,
Sari
FR
, et al
Candesartan cilexetil protects from cardiac myosin induced cardiotoxicity via reduction of endoplasmic reticulum stress and apoptosis in rats: involvement of ACE2-Ang (1-7)-mas axis
.
Toxicology
2012
;
291
:
139
45
.
19.
Zagar
TM
,
Cardinale
DM
,
Marks
LB
. 
Breast cancer therapy-associated cardiovascular disease
.
Nat Rev Clin Oncol
2016
;
13
:
172
84
.
20.
Zamorano
JL
,
Lancellotti
P
,
Rodriguez Muñoz
D
,
Aboyans
V
,
Asteggiano
R
,
Galderisi
M
, et al
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
:
2768
801
.
21.
Scholz-Kreisel
P
,
Spix
C
,
Blettner
M
,
Eckerle
S
,
Faber
J
,
Wild
P
, et al
Prevalence of cardiovascular late sequelae in long-term survivors of childhood cancer: a systematic review and meta-analysis
.
Pediatr Blood Cancer
2017
;
64
.
22.
Cardinale
D
,
Colombo
A
,
Bacchiani
G
,
Tedeschi
I
,
Meroni
CA
,
Veglia
F
, et al
Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy
.
Circulation
2015
;
131
:
1981
8
.
23.
Sodergren
SC
,
Copson
E
,
White
A
,
Efficace
F
,
Sprangers
M
,
Fitzsimmons
D
, et al
Systematic review of the side effects associated with anti-HER2-Targeted therapies used in the treatment of breast cancer, on behalf of the EORTC quality of life group
.
Target Oncol
2016
;
11
:
277
92
.
24.
Bowles
EJA
,
Wellman
R
,
Feigelson
HS
,
Onitilo
AA
,
Freedman
AN
,
Delate
T
, et al
Risk of heart failure in breast cancer patients after anthracycline and trastuzumab treatment: a retrospective cohort study
.
J Natl Cancer Inst
2012
;
104
:
1293
305
.
25.
Zeglinski
M
,
Ludke
A
,
Jassal
DS
,
Singal
PK
. 
Trastuzumab-induced cardiac dysfunction: A “dual-hit
”.
Exp Clin Cardiol
2011
;
16
:
70
4
.
26.
Vermeulen
Z
,
Segers
VFM
,
De Keulenaer
GW
. 
ErbB2 signaling at the crossing between heart failure and cancer
.
Basic Res Cardiol
2016
;
111
:
1
14
.
27.
Harbeck
N
,
Ewer
MS
,
De Laurentiis
M
,
Suter
TM
,
Ewer
SM
. 
Cardiovascular complications of conventional and targeted adjuvant breast cancer therapy
.
Ann Oncol Off J Eur Soc Med Oncol
2011
;
22
:
1250
8
.
28.
Pondé
NF
,
Lambertini
M
,
de Azambuja
E
. 
Twenty years of anti-HER2 therapy-associated cardiotoxicity
.
ESMO Open
2016
;
1
:
e000073
.
29.
Ganz
PA
,
Romond
EH
,
Cecchini
RS
,
Rastogi
P
,
Geyer
CE
,
Swain
SM
, et al
Long-term follow-up of cardiac function and quality of life for patients in NSABP protocol B-31/NRG oncology: a randomized trial comparing the safety and efficacy of doxorubicin and cyclophosphamide (AC) followed by paclitaxel with AC followed by paclitax
.
J Clin Oncol
2017
;
35
:
3942
8
.
30.
Patel
VB
,
Lezutekong
JN
,
Chen
X
,
Oudit
GY
. 
Recombinant human ACE2 and the angiotensin 1-7 axis as potential new therapies for heart failure
.
Can J Cardiol
2017
;
33
:
943
6
.
31.
Akolkar
G
,
Bhullar
N
,
Bews
H
,
Shaikh
B
,
Premecz
S
,
Bordun
K-A
, et al
The role of renin angiotensin system antagonists in the prevention of doxorubicin and trastuzumab induced cardiotoxicity
.
Cardiovasc Ultrasound
2015
;
13
:
18
.
32.
Monti
M
,
Terzuoli
E
,
Ziche
M
,
Morbidelli
L
. 
The sulphydryl containing ACE inhibitor Zofenoprilat protects coronary endothelium from Doxorubicin-induced apoptosis
.
Pharmacol Res
2013
;
76
:
171
81
.
33.
Sacco
G
,
Bigioni
M
,
Evangelista
S
,
Goso
C
,
Manzini
S
,
Maggi
CA
. 
Cardioprotective effects of zofenopril, a new angiotensin-converting enzyme inhibitor, on doxorubicin-induced cardiotoxicity in the rat
.
Eur J Pharmacol
2001
;
414
:
71
8
.
34.
Toko
H
,
Oka
T
,
Zou
Y
,
Sakamoto
M
,
Mizukami
M
,
Sano
M
, et al
Angiotensin II type 1a receptor mediates doxorubicin-induced cardiomyopathy
.
Hypertens Res
2002
;
25
:
597
603
.
35.
Rashikh
A
,
Pillai
KK
,
Najmi
AK
. 
Protective effect of a direct renin inhibitor in acute murine model of cardiotoxicity and nephrotoxicity
.
Fundam Clin Pharmacol
2014
;
28
:
489
500
.
36.
Al-Harbi
NO
,
Imam
F
,
Nadeem
A
,
Al-Harbi
MM
,
Iqbal
M
,
Rahman
S
, et al
Protection against tacrolimus-induced cardiotoxicity in rats by olmesartan and aliskiren
.
Toxicol Mech Methods
2014
;
24
:
697
702
.
37.
Nakamae
H
,
Tsumura
K
,
Terada
Y
,
Nakane
T
,
Nakamae
M
,
Ohta
K
, et al
Notable effects of angiotensin II receptor blocker, valsartan, on acute cardiotoxic changes after standard chemotherapy with cyclophosphamide, doxorubicin, vincristine, and prednisolone
.
Cancer
2005
;
104
:
2492
8
.
38.
Georgakopoulos
P
,
Roussou
P
,
Matsakas
E
,
Karavidas
A
,
Anagnostopoulos
N
,
Marinakis
T
, 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
:
894
6
.
39.
Cadeddu
C
,
Piras
A
,
Mantovani
G
,
Deidda
M
,
Dessì
M
,
Madeddu
C
, et al
Protective effects of the angiotensin II receptor blocker telmisartan on epirubicin-induced inflammation, oxidative stress, and early ventricular impairment
.
Am Heart J
2010
;
160
:
487.e1
7
.
40.
Dessì
M
,
Piras
A
,
Madeddu
C
,
Cadeddu
C
,
Deidda
M
,
Massa
E
, et al
Long-term protective effects of the angiotensin receptor blocker telmisartan on epirubicin-induced inflammation, oxidative stress and myocardial dysfunction
.
Exp Ther Med
2011
;
2
:
1003
9
.
41.
Liu
L
,
Liu
Z
,
Liu
Y
,
Zheng
Z
,
Liang
X
,
Han
Y
, et al
[Preventive effect of low-dose carvedilol combined with candesartan on the cardiotoxicity of anthracycline drugs in the adjuvant chemotherapy of breast cancer]
.
Zhonghua Zhong Liu Za Zhi
2013
;
35
:
936
40
.
42.
Bosch
X
,
Rovira
M
,
Sitges
M
,
Domènech
A
,
Ortiz-Pérez
JT
,
de Caralt
TM
, 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
:
2355
62
.
43.
Radulescu
D
,
Buzdugan
E
,
Ciuleanu
TE
,
Todor
N
,
Stoicescu
L
. 
Can the epirubicin cardiotoxicity in cancer patients be prevented by angiotensin converting enzyme inhibitors? J BUON
2013
;
18
:
1052
7
.
44.
Janbabai
G
,
Nabati
M
,
Faghihinia
M
,
Azizi
S
,
Borhani
S
,
Yazdani
J
. 
Effect of enalapril on preventing anthracycline-induced cardiomyopathy
.
Cardiovasc Toxicol
2017
;
17
:
130
9
.
45.
Gulati
G
,
Heck
SL
,
Ree
AH
,
Hoffmann
P
,
Schulz-Menger
J
,
Fagerland
MW
, et al
Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): a 2 × 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol
.
Eur Heart J
2016
;
37
:
1671
80
.
46.
Boekhout
AH
,
Gietema
JA
,
Milojkovic Kerklaan
B
,
van Werkhoven
ED
,
Altena
R
,
Honkoop
A
, et al
Angiotensin II-receptor inhibition with candesartan to prevent trastuzumab-related cardiotoxic effects in patients with early breast cancer: a randomized clinical trial
.
JAMA Oncol
2016
;
2
:
1030
7
.
47.
Pituskin
E
,
Mackey
JR
,
Koshman
S
,
Jassal
D
,
Pitz
M
,
Haykowsky
MJ
, et al
Multidisciplinary approach to novel therapies in cardio-oncology research (MANTICORE 101-Breast): a randomized trial for the prevention of trastuzumab-associated cardiotoxicity
.
J Clin Oncol
2017
;
35
:
870
7
.
48.
Silber
JH
,
Cnaan
A
,
Clark
BJ
,
Paridon
SM
,
Chin
AJ
,
Rychik
J
, et al
Enalapril to prevent cardiac function decline in long-term survivors of pediatric cancer exposed to anthracyclines
.
J Clin Oncol
2004
;
22
:
820
8
.
49.
Cardinale
D
,
Colombo
A
,
Sandri
MT
,
Lamantia
G
,
Colombo
N
,
Civelli
M
, et al
Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition
.
Circulation
2006
;
114
:
2474
81
.
50.
Gujral
DM
,
Lloyd
G
,
Bhattacharyya
S
. 
Effect of prophylactic betablocker or ACE inhibitor on cardiac dysfunction & heart failure during anthracycline chemotherapy ± trastuzumab
.
Breast
2018
;
37
:
64
71
.
51.
Yancy
CW
,
Jessup
M
,
Bozkurt
B
,
Butler
J
,
Casey
DE
,
Colvin
MM
, et al
2016 ACC/AHA/HFSA focused update on new pharmacological therapy for heart failure: an update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines and the Heart Failure Society of America
.
J Am Coll Cardiol
2016
;
68
:
1476
88
.
52.
Ponikowski
P
,
Voors
AA
,
Anker
SD
,
Bueno
H
,
Cleland
JGF
,
Coats
AJS
, et al
2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure
.
Eur Heart J
2016
;
37
:
2129
200
.
53.
Cardinale
D
,
Colombo
A
,
Lamantia
G
,
Colombo
N
,
Civelli
M
,
De Giacomi
G
, et al
Anthracycline-induced cardiomyopathy: clinical relevance and response to pharmacologic therapy
.
J Am Coll Cardiol
2010
;
55
:
213
20
.
54.
Wittayanukorn
S
,
Qian
J
,
Westrick
SC
,
Billor
N
,
Johnson
B
,
Hansen
RA
. 
Prevention of trastuzumab and anthracycline-induced cardiotoxicity using angiotensin-converting enzyme inhibitors or β-blockers in older adults with breast cancer
.
Am J Clin Oncol
2017
;
0
:
1
10
.
55.
Armenian
SH
,
Lacchetti
C
,
Barac
A
,
Carver
J
,
Constine
LS
,
Denduluri
N
, 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
:
893
911
.
56.
Curigliano
G
,
Cardinale
D
,
Suter
T
,
Plataniotis
G
,
de Azambuja
E
,
Sandri
MT
, et al
Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO clinical practice guidelines
.
Ann Oncol
2012
;
23
:
vii155
66
.
57.
Tocchetti
CG
,
Ragone
G
,
Coppola
C
,
Rea
D
,
Piscopo
G
,
Scala
S
, et al
Detection, monitoring, and management of trastuzumab-induced left ventricular dysfunction: an actual challenge
.
Eur J Heart Fail
2012
;
14
:
130
7
.
58.
Lipshultz
SE
,
Franco
VI
,
Miller
TL
,
Colan
SD
,
Sallan
SE
. 
Cardiovascular disease in adult survivors of childhood cancer
.
Annu Rev Med
2015
;
66
:
161
76
.
59.
Rosenkranz
S
. 
TGF-beta1 and angiotensin networking in cardiac remodeling
.
Cardiovasc Res
2004
;
63
:
423
32
.
60.
de Jesus-Gonzalez
N
,
Robinson
E
,
Moslehi
J
,
Humphreys
BD
. 
Management of antiangiogenic therapy-induced hypertension
.
Hypertension
2012
;
60
:
607
15
.
61.
Ravaud
A
,
Schmidinger
M
. 
Clinical biomarkers of response in advanced renal cell carcinoma
.
Ann Oncol
2013
;
24
:
2935
42
.
62.
Robinson
ES
,
Khankin
EV
,
Karumanchi
SA
,
Humphreys
BD
. 
Hypertension induced by vascular endothelial growth factor signaling pathway inhibition: mechanisms and potential use as a biomarker
.
Semin Nephrol
2010
;
30
:
591
601
.
63.
Ghatalia
P
,
Morgan
CJ
,
Je
Y
,
Nguyen
PL
,
Trinh
Q-D
,
Choueiri
TK
, et al
Congestive heart failure with vascular endothelial growth factor receptor tyrosine kinase inhibitors
.
Crit Rev Oncol Hematol
2015
;
94
:
228
37
.
64.
Small
HY
,
Montezano
AC
,
Rios
FJ
,
Savoia
C
,
Touyz
RM
. 
Hypertension due to antiangiogenic cancer therapy with vascular endothelial growth factor inhibitors: understanding and managing a new syndrome
.
Can J Cardiol
2014
;
30
:
534
43
.
65.
Kappers
MHW
,
van Esch
JHM
,
Sluiter
W
,
Sleijfer
S
,
Danser
AHJ
,
van den Meiracker
AH
. 
Hypertension induced by the tyrosine kinase inhibitor sunitinib is associated with increased circulating endothelin-1 levels
.
Hypertension
2010
;
56
:
675
81
.
66.
Izzedine
H
,
Massard
C
,
Spano
JP
,
Goldwasser
F
,
Khayat
D
,
Soria
JC
. 
VEGF signalling inhibition-induced proteinuria: mechanisms, significance and management
.
Eur J Cancer
2010
;
46
:
439
48
.
67.
Penttilä
P
,
Rautiola
J
,
Poussa
T
,
Peltola
K
,
Bono
P
. 
Angiotensin Inhibitors as treatment of sunitinib/pazopanib-induced hypertension in metastatic renal cell carcinoma
.
Clin Genitourin Cancer
2017
;
15
:
384
90
.
e3
.
68.
Emile
G
,
Pujade-Lauraine
E
,
Alexandre
J
. 
Should we use the angiotensin-converting enzyme inhibitors for the treatment of anti-VEGF-induced hypertension?
Ann Oncol
2014
;
25
:
1669
70
.
69.
Abd-Alhaseeb
MM
,
Zaitone
SA
,
Abou-El-Ela
SH
,
Moustafa
YM
. 
Olmesartan potentiates the anti-angiogenic effect of sorafenib in mice bearing Ehrlich's ascites carcinoma: role of angiotensin (1-7)
.
PLoS One
2014
;
9
:
e85891
.
70.
Koukourakis
MI
. 
Radiation damage and radioprotectants: new concepts in the era of molecular medicine
.
Br J Radiol
2012
;
85
:
313
30
.
71.
Tomic
R
,
Jacobs
E
,
Medhora
M
,
Antonescu-turcu
A
,
Swarajit
G
. 
Role of renin–angiotensin system in radiation-induced lung injury
.
Chest
2008
;
134
:
p127001
.
72.
Cao
S
,
Wu
R
. 
Expression of angiotensin II and aldosterone in radiation-induced lung injury
.
Cancer Biol Med
2012
;
9
:
254
60
.
73.
Jaggi
JS
,
Seshan
SV
,
McDevitt
MR
,
Sgouros
G
,
Hyjek
E
,
Scheinberg
DA
. 
Mitigation of radiation nephropathy after internal alpha-particle irradiation of kidneys
.
Int J Radiat Oncol Biol Phys
2006
;
64
:
1503
12
.
74.
Mahmood
J
,
Jelveh
S
,
Zaidi
A
,
Doctrow
SR
,
Medhora
M
,
Hill
RP
. 
Targeting the renin–angiotensin system combined with an antioxidant is highly effective in mitigating radiation-induced lung damage
.
Int J Radiat Oncol Biol Phys
2014
;
89
:
722
8
.
75.
Robbins
ME
,
Zhao
W
,
Garcia-Espinosa
MA
,
Diz
DI
. 
Renin-angiotensin system blockers and modulation of radiation-induced brain injury
.
Curr Drug Targets
2010
;
11
:
1413
22
.
76.
Authors on behalf of ICRP
,
Stewart
FA
,
Akleyev
AV
,
Hauer-Jensen
M
,
Hendry
JH
,
Kleiman
NJ
, et al
ICRP publication 118: ICRP statement on tissue reactions and early and late effects of radiation in normal tissues and organs–threshold doses for tissue reactions in a radiation protection context
.
Ann ICRP
2012
;
41
:
1
322
.
77.
Kohl
RR
,
Kolozsvary
A
,
Brown
SL
,
Zhu
G
,
Kim
JH
. 
Differential radiation effect in tumor and normal tissue after treatment with ramipril, an angiotensin-converting enzyme inhibitor
.
Radiat Res
2007
;
168
:
440
5
.
78.
Willey
JS
,
Bracey
DN
,
Gallagher
PE
,
Tallant
EA
,
Wiggins
WF
,
Callahan
MF
, et al
Angiotensin-(1-7) attenuates skeletal muscle fibrosis and stiffening in a mouse model of extremity sarcoma radiation therapy
.
J Bone Joint Surg Am
2016
;
98
:
48
55
.
79.
Moore
ED
,
Kooshki
M
,
Metheny-Barlow
LJ
,
Gallagher
PE
,
Robbins
ME
. 
Angiotensin-(1-7) prevents radiation-induced inflammation in rat primary astrocytes through regulation of MAP kinase signaling
.
Free Radic Biol Med
2013
;
65
:
1060
8
.
80.
Rodgers
KE
,
Xiong
S
,
DiZerega
GS
. 
Accelerated recovery from irradiation injury by angiotensin peptides
.
Cancer Chemother Pharmacol
2002
;
49
:
403
11
.
81.
Alite
F
,
Balasubramanian
N
,
Adams
W
,
Surucu
M
,
Mescioglu
I
,
Harkenrider
MM
. 
Decreased risk of radiation pneumonitis with coincident concurrent use of angiotensin-converting enzyme inhibitors in patients receiving lung stereotactic body radiation therapy
.
Am J Clin Oncol
2018
;
41
:
576
80
.
82.
Bracci
S
,
Valeriani
M
,
Agolli
L
,
De Sanctis
V
,
Maurizi Enrici
R
,
Osti
MF
. 
Renin-angiotensin system inhibitors might help to reduce the development of symptomatic radiation pneumonitis after stereotactic body radiotherapy for lung cancer
.
Clin Lung Cancer
2016
;
17
:
189
97
.
83.
Harder
EM
,
Park
HS
,
Nath
SK
,
Mancini
BR
,
Decker
RH
. 
Angiotensin-converting enzyme inhibitors decrease the risk of radiation pneumonitis after stereotactic body radiation therapy
.
Pract Radiat Oncol
2015
;
5
:
e643
9
.
84.
Jenkins
P
,
Welsh
A
. 
Computed tomography appearance of early radiation injury to the lung: correlation with clinical and dosimetric factors
.
Int J Radiat Oncol Biol Phys
2011
;
81
:
97
103
.
85.
Kharofa
J
,
Cohen
EP
,
Tomic
R
,
Xiang
Q
,
Gore
E
. 
Decreased risk of radiation pneumonitis with incidental concurrent use of angiotensin-converting enzyme inhibitors and thoracic radiation therapy
.
Int J Radiat Oncol Biol Phys
2012
;
84
:
238
43
.
86.
Kharofa
J
,
Gore
E
. 
Symptomatic radiation pneumonitis in elderly patients receiving thoracic irradiation
.
Clin Lung Cancer
2013
;
14
:
283
7
.
87.
Wang
H
,
Liao
Z
,
Zhuang
Y
,
Xu
T
,
Nguyen
Q-N
,
Levy
LB
, et al
Do angiotensin-converting enzyme inhibitors reduce the risk of symptomatic radiation pneumonitis in patients with non-small cell lung cancer after definitive radiation therapy? Analysis of a single-institution database
.
Int J Radiat Oncol Biol Phys
2013
;
87
:
1071
7
.
88.
Wang
LW
,
Fu
XL
,
Clough
R
,
Sibley
G
,
Fan
M
,
Bentel
GC
, et al
Can angiotensin-converting enzyme inhibitors protect against symptomatic radiation pneumonitis?
Radiat Res
2000
;
153
:
405
10
.
89.
Small
W
,
James
JL
,
Moore
TD
,
Fintel
DJ
,
Lutz
ST
,
Movsas
B
, et al
Utility of the ACE inhibitor captopril in mitigating radiation-associated pulmonary toxicity in lung cancer: results from NRG oncology RTOG 0123
.
Am J Clin Oncol
2018
;
41
:
396
401
.
90.
Alashkham
A
,
Paterson
C
,
Rauchhaus
P
,
Nabi
G
. 
Can angiotensin-converting enzyme inhibitors reduce the incidence, severity, and duration of radiation proctitis?
Int J Radiat Oncol Biol Phys
2016
;
94
:
93
101
.
91.
Wedlake
LJ
,
Silia
F
,
Benton
B
,
Lalji
A
,
Thomas
K
,
Dearnaley
DP
, et al
Evaluating the efficacy of statins and ACE-inhibitors in reducing gastrointestinal toxicity in patients receiving radiotherapy for pelvic malignancies
.
Eur J Cancer
2012
;
48
:
2117
24
.
92.
Cohen
EP
,
Bedi
M
,
Irving
AA
,
Jacobs
E
,
Tomic
R
,
Klein
J
, et al
Mitigation of late renal and pulmonary injury after hematopoietic stem cell transplantation
.
Int J Radiat Oncol Biol Phys
2012
;
83
:
292
6
.
93.
Jaworski
C
,
Mariani
JA
,
Wheeler
G
,
Kaye
DM
. 
Cardiac complications of thoracic irradiation
.
J Am Coll Cardiol
2013
;
61
:
2319
28
.
94.
Carpentier
AF
,
Ferrari
D
,
Bailon
O
,
Ursu
R
,
Banissi
C
,
Dubessy
A-L
, et al
Steroid-sparing effects of angiotensin-II inhibitors in glioblastoma patients
.
Eur J Neurol
2012
;
19
:
1337
42
.
95.
Kourilsky
A
,
Bertrand
G
,
Ursu
R
,
Doridam
J
,
Barlog
C
,
Faillot
T
, et al
Impact of angiotensin-II receptor blockers on vasogenic edema in glioblastoma patients
.
J Neurol
2016
;
263
:
524
30
.
96.
Chowdhary
M
,
Okwan-Duodu
D
,
Switchenko
JM
,
Press
RH
,
Jhaveri
J
,
Buchwald
ZS
, et al
Angiotensin receptor blockade: a novel approach for symptomatic radiation necrosis after stereotactic radiosurgery
.
J Neurooncol
2018
;
136
:
289
98
.
97.
Conner
KR
,
Payne
VS
,
Forbes
ME
,
Robbins
ME
,
Riddle
DR
. 
Effects of the AT1 receptor antagonist L-158,809 on microglia and neurogenesis after fractionated whole-brain irradiation
.
Radiat Res
2010
;
173
:
49
61
.
98.
Evans
WJ
,
Morley
JE
,
Argilés
J
,
Bales
C
,
Baracos
V
,
Guttridge
D
, et al
Cachexia: a new definition
.
Clin Nutr
2008
;
27
:
793
9
.
99.
Fearon
K
,
Strasser
F
,
Anker
SD
,
Bosaeus
I
,
Bruera
E
,
Fainsinger
RL
, et al
Definition and classification of cancer cachexia: an international consensus
.
Lancet Oncol
2011
;
12
:
489
95
.
100.
Muscaritoli
M
,
Molfino
A
,
Lucia
S
,
Rossi Fanelli
F
. 
Cachexia: a preventable comorbidity of cancer. A T.A.R.G.E.T. approach
.
Crit Rev Oncol Hematol
2015
;
94
:
251
9
.
101.
Ali
S
,
Garcia
JM
. 
Sarcopenia, cachexia and aging: diagnosis, mechanisms and therapeutic options—a mini-review
.
Gerontology
2014
;
60
:
294
305
.
102.
Bruera
E
. 
ABC of palliative care. Anorexia, cachexia, and nutrition
.
BMJ
1997
;
315
:
1219
22
.
103.
Dalla Libera
L
,
Ravara
B
,
Angelini
A
,
Rossini
K
,
Sandri
M
,
Thiene
G
, et al
Beneficial effects on skeletal muscle of the angiotensin II type 1 receptor blocker irbesartan in experimental heart failure
.
Circulation
2001
;
103
:
2195
200
.
104.
Onder
G
,
Penninx
BWJH
,
Balkrishnan
R
,
Fried
LP
,
Chaves
PHM
,
Williamson
J
, et al
Relation between use of angiotensin-converting enzyme inhibitors and muscle strength and physical function in older women: an observational study
.
Lancet
2002
;
359
:
926
30
.
105.
Chen
S-Z
,
Xiao
J-D
. 
Rosiglitazone and imidapril alone or in combination alleviate muscle and adipose depletion in a murine cancer cachexia model
.
Tumour Biol
2014
;
35
:
323
32
.
106.
Murphy
KT
,
Chee
A
,
Trieu
J
,
Naim
T
,
Lynch
GS
. 
Inhibition of the renin-angiotensin system improves physiological outcomes in mice with mild or severe cancer cachexia
.
Int J cancer
2013
;
133
:
1234
46
.
107.
Springer
J
,
Tschirner
A
,
Haghikia
A
,
von Haehling
S
,
Lal
H
,
Grzesiak
A
, et al
Prevention of liver cancer cachexia-induced cardiac wasting and heart failure
.
Eur Heart J
2014
;
35
:
932
41
.
108.
Wang
C
,
Guo
D
,
Wang
Q
,
You
S
,
Qiao
Z
,
Liu
Y
, et al
Aliskiren targets multiple systems to alleviate cancer cachexia
.
Oncol Rep
2016
;
36
:
3014
22
.
109.
Roldan
CJ
,
Song
J
,
Engle
MP
,
Dougherty
PM
. 
Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers modulate the function of myelinated fibers after chemotherapy: a quantitative sensory testing study
.
Pain Phys
2017
;
20
:
281
92
.
110.
Komaki
K
,
Kusaba
T
,
Tanaka
M
,
Kado
H
,
Shiotsu
Y
,
Matsui
M
, et al
Lower blood pressure and risk of cisplatin nephrotoxicity: a retrospective cohort study
.
BMC Cancer
2017
;
17
:
144
.