Next Article in Journal
Correction: Iwan et al. Pro-Oxidative Effect of KIO3 and Protective Effect of Melatonin in the Thyroid—Comparison to Other Tissues. Life 2021, 11, 592
Next Article in Special Issue
Valvular Heart Disease following Anthracycline Therapy—Is It Time to Look beyond Ejection Fraction?
Previous Article in Journal
Intraoperative Circulatory Support in Lung Transplantation: Current Trend and Its Evidence
Previous Article in Special Issue
Cardiac Events in Childhood Cancer Survivors Treated with Anthracyclines: The Value of Previous Myocardial Strain Measurement
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardio-Oncology Rehabilitation—Present and Future Perspectives

1
The Department of Cardiology, Rambam Health Care Campus, Haifa 3109601, Israel
2
Technion—The Ruth and Bruce Rappaport Faculty of Medicine, Haifa 3525433, Israel
*
Author to whom correspondence should be addressed.
Life 2022, 12(7), 1006; https://doi.org/10.3390/life12071006
Submission received: 6 May 2022 / Revised: 24 June 2022 / Accepted: 28 June 2022 / Published: 7 July 2022

Abstract

:
Recent advances in cancer therapy have led to increased survival rates for cancer patients, but also allowed cardiovascular complications to become increasingly evident, with more than 40% of cancer deaths now being attributed to cardiovascular diseases. Cardiotoxicity is the most concerning cardiovascular complication, one caused mainly due to anti-cancer drugs. Among the harmful mechanisms of these drugs are DNA damage, endothelial dysfunction, and oxidative stress. Cancer patients can suffer reduced cardiorespiratory fitness as a secondary effect of anti-cancer therapies, tumor burden, and deconditioning. In the general population, regular exercise can reduce the risk of cardiovascular morbidity, mortality, and cancer. Exercise-induced modifications of gene expression result in improvements of cardiovascular parameters and an increased general fitness, influencing telomere shortening, oxidative stress, vascular function, and DNA repair mechanisms. In cancer patients, exercise training is generally safe and well-tolerated; it is associated with a 10–15% improvement in cardiorespiratory fitness and can potentially counteract the adverse effects of anti-cancer therapy. It is well known that exercise programs can benefit patients with heart disease and cancer, but little research has been conducted with cardio-oncology patients. To date, there are a limited number of effective protective treatments for preventing or reversing cardiotoxicity caused by cancer therapy. Cardiac rehabilitation has the potential to mitigate cardiotoxicity based on the benefits already proven in populations suffering from either cancer or heart diseases. Additionally, the fact that cardiotoxic harm mechanisms coincide with similar mechanisms positively affected by cardiac rehabilitation makes cardiac rehabilitation an even more plausible option for cardio-oncology patients. Due to unstable functional capacity and fluctuating immunocompetence, these patients require specially tailored exercise programs designed collaboratively by cardiologists and oncologists. As the digital era is here, with the digital world and the medical world continuously intertwining, a remote, home-based cardio-oncology rehabilitation program may be a solution for this population.

1. Introduction

Recent cancer treatment advances have yielded tremendous gains, leading to longer survival and better quality of life [1]. Cancer death rates in the United States decreased by 20% from 1991 (215.1 per 100,000 in the general population) to 2010 (171.8 per 100,000 in the general population), leading to millions of cancer survivors [1]. These advances are derived from early cancer diagnoses and generally improved treatments. More than 60 anti-cancer drugs have been approved by the FDA in recent years. In addition, a deeper understanding of tumor biology has led to an entire range of new, molecularly targeted drugs. The strategy of combining different types of therapy has also shown powerful results [1].
The advances in cancer therapy have led to increased survival rates of cancer patients but also allowed for short- and long-term cardiovascular complications to become evident, leading, in turn, to premature morbidity and mortality among cancer survivors [2]. In recent years, many of the deaths among cancer patients were not attributed directly to the index-cancer and more than 40% of cancer patients’ deaths were attributed to cardiovascular diseases [3].
In general, cardiovascular complications of cancer therapy can be divided into nine main categories: myocardial dysfunction and heart failure (HF); coronary artery disease (CAD); valvular disease; arrhythmias; arterial hypertension; thromboembolic disease; peripheral vascular disease and stroke; pulmonary hypertension; and pericardial complications [2].
Cancer-therapy-related cardiac dysfunction is increasingly evident within the expanding use of novel chemotherapeutic agents. This dysfunction is a serious complication that could eventually culminate in the development of life-threatening myocardial dysfunction and HF [4], and it is one of the most common causes of non-cancer deaths [3]. HF is typically difficult to treat, substantially affecting overall morbidity and mortality, and requiring long-term therapy. Myocardial dysfunction and HF, frequently described as cardiotoxicity, are the most concerning cardiovascular complications. Cardiotoxicity may involve direct effects of the cancer treatment on the function and structure of the heart, or it may be due to accelerated developments in cardiovascular diseases (CVD) [2].
Among the leading causes for cardiotoxicity are anti-cancer drugs and radiotherapy. Anthracycline-based therapy can result in irreversible cardiac damage and HF, with an incidence of 5–48%, in a dose-dependent manner [2]. The mechanisms of 5-fluorouracil induced myocardial ischemia are multifactorial and include coronary vasospasm and endothelial injury. Other conventional chemotherapies, including Cyclophosphamide, Cisplatin, Ifosfamide, and Taxanes can lead to cardiotoxicities as well. These severe adverse consequences of therapy are particularly relevant for young cancer survivors who display increased rates of cardiovascular diseases, even several decades after therapy [2]. Immunotherapies and targeted therapies, such as Trastuzumab and other therapies targeting anti-human epidermal growth factor receptor 2, may cause cardiac dysfunction as well, especially with concomitant or previous use of anthracyclines. Vascular endothelial growth factor inhibitors can cause both reversible and irreversible cardiac side effects (particularly when used with or after conventional chemotherapies) and arterial hypertension, potentially affecting cardiac function. Potent inhibitors of BCR-ABL have also demonstrated an association with cardiovascular events. Proteasome inhibitors for treatment of multiple myeloma can potentially cause cardiac dysfunction as well. Among survivors exposed to radiotherapy, the risk of HF was increased 4.9-fold, and marked interstitial myocardial fibrosis is common in radiotherapy-induced cardiotoxicity. HF may also be aggravated by concomitant radiation-induced valvular heart disease and coronary artery disease (CAD) [2].
Risk factors for cardiotoxicity include current HF, asymptomatic left ventricle dysfunction (ejection fracture less than 50%), CAD, valvular heart disease, hypertensive heart or left ventricle hypertrophy, cardiomyopathy, arrhythmias, prior cardiotoxicity (anthracyclines, mediastinal radiotherapy, or chest radiotherapy), age of less than 18 years old or more than 50 years old, family history of CAD, and traditional cardiovascular co-morbidities and lifestyle risk factors, such as hypertension, diabetes, and hyperlipidemia [2].

2. Non-Cardiac Physical Deconditioning in Cancer Patients

Most cancer patients and cancer survivors will experience compromised physical and cognitive functioning due to cancer treatments and their side effects, resulting in a negative impact on their quality of life and survival rates [5]. Reduced cardiorespiratory fitness (CRF) in cancer patients is multifactorial. Cancer patients are often older than the general population and commonly present with pulmonary, cardiovascular, and/or musculoskeletal complications. Treatment with anti-cancer therapies, combined with tumor burden, can simultaneously impact components in the oxygen cascade, including the pulmonary, cardiac, hematological, vascular, and musculoskeletal systems. Indirect effects of physical inactivity (deconditioning) will further contribute to reduced CRF [6]. These patients also develop multiple medical conditions, often related to the late and long-term effects of their cancer treatment, as well as conditions related to premature aging [7]. Several studies following long-term cancer survivors have shown significant impairment in activities, physical limitations [8,9,10], and decreased exercise capacity in different cancer populations, including chemotherapy-treated and post-surgical populations [11,12,13].
The main risk factors for physical deconditioning in cancer patients include the attributes of female gender, lower income and education, central nervous system and bone cancers, brain radiotherapy, and premature gonadal failure (caused by androgen deprivation, alkylating agents, and pelvic irradiation) [11].

3. Mechanisms of Cardiotoxicity

There are several theories regarding the different mechanisms for cancer treatment cardiotoxicity. Contemporary treatment methods are effective in treating different modalities of cancers; however, these reagents act through interference with DNA replication or by preventing DNA repair, causing endothelial dysfunction, generating reactive oxygen species, or eliciting non-specific immune responses [14]. Anthracyclines have become a central chemotherapeutic treatment option because of their high efficacy [15]. Anthracycline cardiotoxicity is estimated to affect 9% of cancer patients treated with anthracyclines [16]. The mechanism is believed to be multifactorial. The two main hypotheses are: (1) oxidative stress, which, in the presence of iron, generates reactive oxygen species that cause lipid peroxidation of the cell membrane, leading to damage of the cardiomyocytes [17,18,19]; and (2) inhibition of topoisomerase IIβ (TOPIIβ), which is active in quiescent nonproliferating cardiomyocytes. The inhibition of TOPIIβ can result in the activation of cell death pathways and inhibition of mitochondrial biogenesis [15,20]. Another suggested mechanism is the dysregulation of cardiomyocyte autophagy [21]. Anthracycline toxicity is dose dependent. Evidence suggests cardiotoxicity occurrences of 10%, 16%, 32%, and 65% at cumulative doxorubicin doses of 250, 300, 400, and 550 mg/m2, respectively [22]. Trastuzumab, functioning as an inhibitor of ErbB2 receptor, interrupts ErbB4/ErbB2 heterodimerization, leading to subsequent mitochondrial energy dysfunction and apoptosis [23,24]. In addition, trastuzumab activates the autophagy-inhibitory Erk/mTOR/Ulk 1 signaling cascade, causing cardiomyocyte apoptosis [25]. 5-fluorouracil (5-FU) can lead to coronary vasospasm through Protein kinase C and Endothelin-I [26,27], direct myocardial injury through alpha-fluoro-beta-alanine (FBAL) (which is a breakdown product of 5-FU [28]), vascular endothelial dysfunction through direct toxic effects, and excess production of reactive oxygen species [29]. Moreover, mitochondrial dysfunction and apoptosis of cardiomyocytes are believed to play an important role in 5-FU induced cardiotoxicity [30]. Cisplatin can induce nuclear and mitochondrial DNA damage [31], as well as the augmentation of oxidative stress and mitochondrial dysfunction [31,32]. Metabolites of cyclophosphamide are responsible for cardiotoxicity due to depletion of antioxidants/ATP level, altered contractility, damaged endothelium, and enhanced pro-inflammatory/pro-apoptotic activities [33]. Tyrosine kinase inhibitor-induced cardiac injury is mediated by oxidative stress, mitochondrial damage, and apoptosis of cardiomyocytes [34]. Immune checkpoint inhibitors, including anti-PD-1, anti-PD-L1, and CTLA-4 blockade, can cause cardiotoxicity as a result of immune inflammation and oxidative stress [30,35,36]. Figure 1 summarizes mechanisms mediating the cardiotoxicity of selected cancer therapeutics. Table 1 details some of the main mechanisms involved in common cancer therapeutics-related cardiotoxicity.

4. Therapies for Prevention of Cardiotoxicity

Drugs such as Dexrazoxane have shown a significant protective effect, but their use is limited, due to their high-cost and the concern about a potential reduction in anti-cancer efficacy and a risk of developing secondary tumors [4]. Apart from Dexrazoxane, the field has thus far focused on utilizing drugs aimed at mitigating maladaptive neurohormonal activation and preventing further deterioration of cardiac function. Conventional HF drugs have demonstrated limited benefit in ameliorating the cardiotoxic damage of chemotherapy. The 2016 ESC guidelines state that the benefits from preventive treatment with ACE inhibitors, ARBs, or beta-blocker therapy remain controversial, and no recommendation can be made at this time [2,39]. Several preliminary studies on animal models found Metformin to exert anti-cancer and cardio-protective properties, although more studies are necessary [40].

5. Exercise Influence on the Cardiovascular System

Regular exercise, in which moderate-to-vigorous intensity is achieved, leads to a reduction in the risks of all-cause mortality, CVD, hypertension, stroke, metabolic syndrome, diabetes, and cancer. Individuals who sustain high exercise levels tend to live longer and have lower mortality rates for both CVD and cancer, compared with the general population [41]. Exercise substantially alters the expression of genes in the human genome. Exercise-induced modifications of gene expression result in rapid, yet transient, improvements in cardiovascular parameters and general fitness [41]. Exercise may prevent telomere shortening (modifying senescence, apoptosis, and oncogenic transformation of cells), affect oxidative stress and vascular function, and induce the upregulation of DNA repair mechanisms [41,42].
Exercise-induced oxidative stress continues to be a controversial topic. In theory, a moderate level of reactive oxygen species (ROS) production during exercise promotes positive physiological adaptation in the active skeletal muscles (e.g., mitochondrial biogenesis, synthesis of antioxidant enzymes, and production of stress proteins), whereas high levels of ROS production result in damage to macromolecular structures (e.g., proteins, lipids, and DNA). The increased risk of CVD occurrence is partially attributed to the development of vascular endothelial dysfunction, while regular endurance exercise is effective for maintaining overall vascular regularity and can prevent a loss of endothelium-dependent vasodilation. It has been shown that training individuals have significantly higher endothelial NOS (eNOS) expression and phospho-eNOS levels, superoxide dismutase, and an improved vascular antioxidant capacity, which was correlated with endothelial function [41]. Past studies have revealed that either endurance exercise or interval training increased the antioxidant capacity of both cardiac and skeletal muscle myocytes. Both short-term (measured in days) and long-term (3 month) endurance exercise training increase antioxidant enzyme activities in the trained muscles and eliminate the contraction-induced oxidative stress that can result from an acute bout of exercise [42]. The reduction in oxidative stress has the potential to reduce the risk of cancer as well. A large epidemiological study involving 1.44 million patients concluded that regular physical activity reduced the risk of 13 different types of cancer [43]. Exercise has been shown to reduce the risk of recurrence of tumor growth as well. The molecular mechanisms responsible for exercise-induced protection against cancer remain unclear, but an upregulation of antioxidant gene expression has been postulated to be a contributing factor [42]. Figure 1 summarizes mechanisms mediating cardiotoxicity, including the beneficial effects of exercise.

6. Rehabilitation for Cancer Patients

The main aims of rehabilitation for cancer patients are to regain physical activity, prevent frailty, and improve aerobic abilities. Aerobic exercise training has been widely established to be one of the most-effective therapies to improve CRF. It improves the reserve capacity of the components of oxygen transport and use, which leads to favorable improvements in VO2peak [6].
Randomized trials indicate that exercise training in cancer patients, following traditional exercise prescription guidelines, is safe and well-tolerated during various conventional therapeutic modalities and is associated with 10–15% improvement in different measures of CRF. Exercise training can negate the adverse effects of therapy upon CRF [6]. Several studies in recent years have found that exercise may attenuate cancer-treatment-induced declines in CRF and improve CRF after the completion of cancer therapy in different cancer populations; the studies also demonstrated the benefit of rehabilitation in high-risk patients (e.g., frail patients, elderly patients, and those undergoing complex surgery) [7]. A meta-analysis of 27 randomized clinical trials of exercise training in cancer patients after the completion of adjuvant therapy showed significantly increased CRF compared with usual care (VO2 peak weighted mean differences, 2.45 mL O2·kg−1·min−1 [95% CI, 1.71–3.19]) [44]. In addition, past trials have even shown correlation with improved cardiovascular outcomes and reduced mortality [44,45,46], although most of those studies are retrospective and include meta-analysis, whereas other trials have found no benefit. Although studies of the effect of exercise on cardiovascular disease outcomes in cancer survivors have focused primarily on CRF, these studies possess a significant clinical importance, because CRF is associated with short- and long-term treatment-related toxicities, symptom burden, and all-cause and cancer-specific mortality [47]. A scientific statement from the American Heart Association in 2019 urged the development of a comprehensive model, as a part of a cardio-oncology rehabilitation program, to identify cancer patients at high risk of CVD or those who have developed cardiotoxicity related to cancer therapies, and to use the multimodality approach of cardiac rehabilitation to prevent or mitigate cardiovascular events [47]. However, there are several important limitations for the participation of cancer patients in rehabilitation programs. Due to treatment side effects, as well as fever and neutropenia during nadir, participation in standard rehabilitation programs at hospital or clinic settings might expose them to various risks and higher rates of complications. In addition, due to changing hemoglobin levels and anemia, it might be difficult to determine and adjust pulse range as an indicator for exercise intensity level. Table 2 summarizes the trial results of the main studies on rehabilitation for cancer patients.

7. Cardiac Rehabilitation in Cardiovascular Patients

The main goals of cardiac rehabilitation are to prevent major adverse cardiovascular events, to improve VO2max, and to prevent mortality. Regular physical activity is an important component of therapy for most cardiovascular diseases (CVD) and is associated with reduced cardiovascular and all-cause mortality. Current guidelines strongly recommend (Class 1, level of evidence A recommendation) that patients with CVD risk factors and an established cardiac disease, including ischemic heart disease (IHD) and HF, should perform a minimum of 150 min of moderate intensity endurance exercise training weekly. Exercise-based cardiac rehabilitation (exCR) reduces cardiac mortality and hospital readmission and improves exercise tolerance and quality of life in different CVD patients [63].
In HF patients, exercise can improve quality of life and fitness. In HF with reduced ejection fraction (HFrEF), different studies have found that exCR improves exercise capacity and quality of life. In the HF-ACTION trial, 2331 patients were randomly assigned to either a monitored aerobic exercise training followed by remote-based exercise or to standard care. After three months, the exercise program group had a statistically significant improvement in the Kansas City Cardiomyopathy Questionnaire compared with the standard care group (mean 5.21 vs. 3.28, p < 0.001) [64]. In addition, while the trial resulted in no significant reductions in all-cause mortality or hospitalization, after correction for prognosis predictors, a significant reduction for either mortality or hospitalization in the exercise group was noted [65]. Several meta-analyses found that exCR can reduce all-cause and HF hospitalizations, especially in highly adherent patients, while the effect on mortality is uncertain [66,67]. In HF with preserved EF (HFpEF), exercise is recommended as well, as it can improve symptoms and exercise capacity. Weight reduction in these patients is extremely important and a positive additional outcome of exercise programs [63,68]. In a meta-analysis including 276 patients with HFpEF, those participating in exercise training had significantly improved cardiorespiratory fitness (mean difference of peak oxygen uptake 2.72 mL/kg per min) and quality of life as assessed by the Minnesota Living With Heart Failure score (mean difference, −3.97) when compared with the control arm [69]. In a randomized control trial including 100 older obese patients with HFpEF, caloric restriction or aerobic exercise training led to increased peak oxygen consumption compared to the control group (1.3 mL/kg per min, p < 0.0011 and 1.2 mL/kg per min, p < 0.001 respectively), with a possible additive effect attained when combining the two interventions [70]. As for HF patients with mildly reduced EF (HFmrEF), present data are scarce, but it is believed that the benefits found in other HF populations should apply [68]. In a recent post hoc analysis on the effect of remote exCR implementation in HF patients, including 195 HFrEF and 381 HFmrEF or HFpEF patients, the completion of a rehabilitation program was a strong negative predictor of death or re-hospitalization in HFrEF (hazard ratio 0.10, p = 0.025) and HFmrEF or HFpEF (hazard ratio 0.11, p = 0.028) patients [71].
Although exCR programs are highly recommended, they are significantly underused. About 10–16% of IHD patients eligible for exCR participate in these programs [72], and this percentage drops to 4.3% when HF patients are involved [73]. ExCR programs are divided into traditional, center-based programs (the prevalent form) and remote, home-based programs (HBCR). One of the advantages of center-based programs is an increase in monitoring capabilities, making such programs suitable for high- and low-risk patients, the frail, and patients who have more severe disease or comorbidities [64], whereas remote programs can better suit patients’ daily routines, thereby potentially increasing their participation rate. Recently, a statement by the American Heart Association determined that new delivery strategies are urgently needed to improve participation, a finding that emphasizes HBCR’s importance [74]. Formerly, studies have found similar clinical outcomes with HBCR compared to center-based programs [74]. HBCR monitoring and communication intensities vary. The spectrum ranges from programs that rely on basic communication, such as telephone calls, to sophisticated telemedicine programs using advanced digital means, including the automatic transfer of exercise data, clinical data, and auto-analysis information to the exCR team.

8. Present Data

Although there is evidence for the beneficial effects of exCR on patients with CVD and on cancer patients, there are very little data on exCR for cardio-oncology patients. Several preventive studies on cancer patients before or during cardiotoxic chemotherapy found that exCR improves hemodynamics, symptoms, and functional disability, but it does not affect cardiotoxicity markers or cardiac function. Kirkham et al. studied the effect of aerobic and resistance training in 37 breast cancer patients during anthracycline therapy. The study showed changes in hemodynamic responses, such as drops in systemic vascular resistance and cardiac output, but no beneficial effect of exercise on cardiac function as assessed by different echocardiography measurements [75]. In a study of 28 breast cancer patients participating in exercise program (2 × 60 min supervised exercise sessions per week) during anthracycline treatment, Howden et al. found that exercise prevented the decline in VO2peak (15 vs. 4% reduction, p = 0.010), but it did not prevent the decline in left ventricle ejection fraction or in the levels of troponin [76]. In another study, Kirkham et al. showed that, in breast cancer patients, 30 min of vigorous-intensity exercise 24 h prior to the first doxorubicin treatment attenuated NT-proBNP release and increased systolic function, but the study included only 24 patients [77]. Searching the literature for studies on the role of exCR in cancer patients with established HF yielded only one trial: a retrospective analysis of 90 patients with cancer who have HF. Exercise programs did not improve any clinical outcomes, including VO2peak, quality of life, hospitalization, and death, but the patients showed no proven association between cancer treatment and HF occurrence [78]. We could not find any trials including patients with HF secondary to cancer treatment toxicity. Cardiac rehabilitation effects are summarized in Table 3.

9. The Gaps

There are certain crucial gaps in the evidence.
To date, there has been no effective treatment to prevent or reverse cancer-therapy-related cardiotoxicity. ExCR cardio-oncology programs are scarce. Specifically, there is a growing need for out-of-hospital programs for cancer patients, as this population suffers intensive hospitalization periods, including an extremely high risk for acquiring infections in the hospital or the clinic, necessitating flexibility in exercise formats tailored to their cancer treatment regimen and disease stage. In addition, there are only limited data regarding exCR for cancer patients with or at risk of cardiotoxicity. The demonstrated benefit of exercise in other clinical populations suggests that exercise may also become a key feature of future programs for oncology patients at risk for cardiotoxicity, or for those who already developed this complication.

10. Future Direction

As exCR programs reduce morbidity and mortality in CVD and improve the fitness and quality of life of cancer patients, further research is urgently needed to elucidate these programs’ effects on cardio-oncology patients.
As these patients are extremely complex, cardiologists and oncologists should create a hybrid rehabilitation program, taking into account different aspects of this unique population. Cardiologists could evaluate for risk factors, including the risk of cardiovascular complications such as arrhythmia and HF, and adjust rehabilitation goals and monitor intensity during exercise accordingly. Oncologists could alert the team to complications and side effects associated with anti-cancer treatments, such as anemia, risk and anticipated timing for neutropenia and risk of infection.
At present, rehabilitation programs’ major drawbacks include low participation rates, a risk negatively influenced during the COVID-19 pandemic. It seems that future cardio-oncology programs will suffer similar limitations, with additional difficulties arising due to patients’ complexity and fear of contagious disease due to immune suppression. A novel approach should be sought to overcome these difficulties. One solution can include a remote, home-based cardio-oncology rehabilitation program with the potential to increase participation rates [74] while reducing the COVID-19 effect and contagion risk.
Currently, in the digital era, the digital world and the medical world continuously intertwine. With the emergence of digital telemedicine capabilities, including arrhythmia monitoring and continuous intra-cardiac measurements [79,80,81], the prospect of remote, digital cardio-oncology programs becoming dominant seems compelling and inevitable. These advanced programs possess the advantage of increased participation, while preserving safety for fragile patients by keeping them out of hospital, thereby saving manpower due to increased efficiency. Different programs should be developed for primary and secondary prevention in cardio-oncology patients, including programs targeting those with and without risk factors for cardiotoxicity, and programs before and after cardiotoxicity treatments, including anti-cancer drugs, radiotherapy, and post-surgery. As remote and digital programs become popular, data will be accumulated, and there will be a need for studies to confirm these programs’ efficacy and safety, and also revealing their effect on prevention and treatment of cardiac dysfunction, quality of life, hospitalization, and mortality.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Masters, G.A.; Krilov, L.; Bailey, H.H.; Brose, M.S.; Burstein, H.; Diller, L.R.; Dizon, D.S.; Fine, H.A.; Kalemkerian, G.P.; Moasser, M.; et al. Clinical cancer advances 2015: Annual report on progress against cancer from the American Society of Clinical Oncology. J. Clin. Oncol. 2015, 33, 786–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Zamorano, J.L.; Lancellotti, P.; Rodriguez Munoz, D.; Aboyans, V.; Asteggiano, R.; Galderisi, M.; Habib, G.; Lenihan, D.J.; Lip, G.Y.H.; Lyon, A.R.; 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, 2768–2801. [Google Scholar] [CrossRef] [PubMed]
  3. Zaorsky, N.G.; Churilla, T.M.; Egleston, B.L.; Fisher, S.G.; Ridge, J.A.; Horwitz, E.M.; Meyer, J.E. Causes of death among cancer patients. Ann. Oncol. 2017, 28, 400–407. [Google Scholar] [CrossRef] [PubMed]
  4. Angsutararux, P.; Luanpitpong, S.; Issaragrisil, S. Chemotherapy-Induced Cardiotoxicity: Overview of the Roles of Oxidative Stress. Oxid. Med. Cell. Longev. 2015, 2015, 795602. [Google Scholar] [CrossRef] [Green Version]
  5. Stout, N.L.; Santa Mina, D.; Lyons, K.D.; Robb, K.; Silver, J.K. A systematic review of rehabilitation and exercise recommendations in oncology guidelines. CA Cancer J. Clin. 2021, 71, 149–175. [Google Scholar] [CrossRef]
  6. Lakoski, S.G.; Eves, N.D.; Douglas, P.S.; Jones, L.W. Exercise rehabilitation in patients with cancer. Nat. Rev. Clin. Oncol. 2012, 9, 288–296. [Google Scholar] [CrossRef] [Green Version]
  7. Kline, R.M.; Arora, N.K.; Bradley, C.J.; Brauer, E.R.; Graves, D.L.; Lunsford, N.B.; McCabe, M.S.; Nasso, S.F.; Nekhlyudov, L.; Rowland, J.H.; et al. Long-Term Survivorship Care after Cancer Treatment-Summary of a 2017 National Cancer Policy Forum Workshop. J. Natl. Cancer Inst. 2018, 110, 1300–1310. [Google Scholar] [CrossRef] [Green Version]
  8. Sweeney, C.; Schmitz, K.H.; Lazovich, D.; Virnig, B.A.; Wallace, R.B.; Folsom, A.R. Functional limitations in elderly female cancer survivors. J. Natl. Cancer Inst. 2006, 98, 521–529. [Google Scholar] [CrossRef] [Green Version]
  9. Robison, L.L.; Green, D.M.; Hudson, M.; Meadows, A.T.; Mertens, A.C.; Packer, R.J.; Sklar, C.A.; Strong, L.C.; Yasui, Y.; Zeltzer, L.K. Long-term outcomes of adult survivors of childhood cancer. Cancer 2005, 104, 2557–2564. [Google Scholar] [CrossRef]
  10. Maunsell, E.; Pogany, L.; Barrera, M.; Shaw, A.K.; Speechley, K.N. Quality of life among long-term adolescent and adult survivors of childhood cancer. J. Clin. Oncol. 2006, 24, 2527–2535. [Google Scholar] [CrossRef]
  11. Stein, K.D.; Syrjala, K.L.; Andrykowski, M.A. Physical and psychological long-term and late effects of cancer. Cancer 2008, 112, 2577–2592. [Google Scholar] [CrossRef] [PubMed]
  12. Jones, L.W.; Watson, D.; Herndon, J.E., 2nd; Eves, N.D.; Haithcock, B.E.; Loewen, G.; Kohman, L. Peak oxygen consumption and long-term all-cause mortality in nonsmall cell lung cancer. Cancer 2010, 116, 4825–4832. [Google Scholar] [CrossRef] [PubMed]
  13. Tonorezos, E.S.; Snell, P.G.; Moskowitz, C.S.; Eshelman-Kent, D.A.; Liu, J.E.; Chou, J.F.; Smith, S.M.; Dunn, A.L.; Church, T.S.; Oeffinger, K.C. Reduced cardiorespiratory fitness in adult survivors of childhood acute lymphoblastic leukemia. Pediatr. Blood Cancer 2013, 60, 1358–1364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Dong, J.; Chen, H. Cardiotoxicity of Anticancer Therapeutics. Front. Cardiovasc. Med. 2018, 5, 9. [Google Scholar] [CrossRef] [Green Version]
  15. Saleh, Y.; Abdelkarim, O.; Herzallah, K.; Abela, G.S. Anthracycline-induced cardiotoxicity: Mechanisms of action, incidence, risk factors, prevention, and treatment. Heart Fail. Rev. 2021, 26, 1159–1173. [Google Scholar] [CrossRef]
  16. Cardinale, D.; Colombo, A.; Bacchiani, G.; Tedeschi, I.; Meroni, C.A.; Veglia, F.; Civelli, M.; Lamantia, G.; Colombo, N.; Curigliano, G.; et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation 2015, 131, 1981–1988. [Google Scholar] [CrossRef] [Green Version]
  17. Simunek, T.; Sterba, M.; Popelova, O.; Adamcova, M.; Hrdina, R.; Gersl, V. Anthracycline-induced cardiotoxicity: Overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacol. Rep. 2009, 61, 154–171. [Google Scholar] [CrossRef]
  18. Suliman, H.B.; Carraway, M.S.; Ali, A.S.; Reynolds, C.M.; Welty-Wolf, K.E.; Piantadosi, C.A. The CO/HO system reverses inhibition of mitochondrial biogenesis and prevents murine doxorubicin cardiomyopathy. J. Clin. Investig. 2007, 117, 3730–3741. [Google Scholar] [CrossRef] [Green Version]
  19. Wallace, K.B. Doxorubicin-induced cardiac mitochondrionopathy. Pharmacol. Toxicol. 2003, 93, 105–115. [Google Scholar] [CrossRef]
  20. Zhang, S.; Liu, X.; Bawa-Khalfe, T.; Lu, L.S.; Lyu, Y.L.; Liu, L.F.; Yeh, E.T. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat. Med. 2012, 18, 1639–1642. [Google Scholar] [CrossRef]
  21. Li, D.L.; Wang, Z.V.; Ding, G.; Tan, W.; Luo, X.; Criollo, A.; Xie, M.; Jiang, N.; May, H.; Kyrychenko, V.; et al. Doxorubicin Blocks Cardiomyocyte Autophagic Flux by Inhibiting Lysosome Acidification. Circulation 2016, 133, 1668–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Bernstein, D. Anthracycline Cardiotoxicity: Worriisome Enough to Have You Quaking? Circ. Res. 2018, 122, 188–190. [Google Scholar] [CrossRef] [PubMed]
  23. Fedele, C.; Riccio, G.; Malara, A.E.; D’Alessio, G.; De Lorenzo, C. Mechanisms of cardiotoxicity associated with ErbB2 inhibitors. Breast Cancer Res. Treat. 2012, 134, 595–602. [Google Scholar] [CrossRef] [PubMed]
  24. Fuller, S.J.; Sivarajah, K.; Sugden, P.H. ErbB receptors, their ligands, and the consequences of their activation and inhibition in the myocardium. J. Mol. Cell. Cardiol. 2008, 44, 831–854. [Google Scholar] [CrossRef] [PubMed]
  25. Mohan, N.; Shen, Y.; Endo, Y.; ElZarrad, M.K.; Wu, W.J. Trastuzumab, but Not Pertuzumab, Dysregulates HER2 Signaling to Mediate Inhibition of Autophagy and Increase in Reactive Oxygen Species Production in Human Cardiomyocytes. Mol. Cancer Ther. 2016, 15, 1321–1331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Mosseri, M.; Fingert, H.J.; Varticovski, L.; Chokshi, S.; Isner, J.M. In vitro evidence that myocardial ischemia resulting from 5-fluorouracil chemotherapy is due to protein kinase C-mediated vasoconstriction of vascular smooth muscle. Cancer Res. 1993, 53, 3028–3033. [Google Scholar]
  27. Thyss, A.; Gaspard, M.H.; Marsault, R.; Milano, G.; Frelin, C.; Schneider, M. Very high endothelin plasma levels in patients with 5-FU cardiotoxicity. Ann. Oncol. 1992, 3, 88. [Google Scholar] [CrossRef]
  28. Muneoka, K.; Shirai, Y.; Yokoyama, N.; Wakai, T.; Hatakeyama, K. 5-Fluorouracil cardiotoxicity induced by alpha-fluoro-beta-alanine. Int. J. Clin. Oncol. 2005, 10, 441–443. [Google Scholar] [CrossRef]
  29. Layoun, M.E.; Wickramasinghe, C.D.; Peralta, M.V.; Yang, E.H. Fluoropyrimidine-Induced Cardiotoxicity: Manifestations, Mechanisms, and Management. Curr. Oncol. Rep. 2016, 18, 35. [Google Scholar] [CrossRef] [Green Version]
  30. Ma, W.; Wei, S.; Zhang, B.; Li, W. Molecular Mechanisms of Cardiomyocyte Death in Drug-Induced Cardiotoxicity. Front. Cell Dev. Biol. 2020, 8, 434. [Google Scholar] [CrossRef]
  31. El-Awady, E.S.E.; Moustafa, Y.M.; Abo-Elmatty, D.M.; Radwan, A. Cisplatin-induced cardiotoxicity: Mechanisms and cardioprotective strategies. Eur. J. Pharmacol. 2011, 650, 335–341. [Google Scholar] [CrossRef] [PubMed]
  32. Demkow, U.; Biatas-Chromiec, B.; Stelmaszczyk-Emmel, A.; Radzikowska, E.; Wiatr, E.; Radwan-Rohrenschef, P.; Szturmowicz, M. The Cardiac Markers and Oxidative Stress Parameters in Advanced Non-Small Cell Lung Cancer Patients Receiving Cisplatin-Based Chemotherapy. EJIFCC 2011, 22, 6–15. [Google Scholar] [PubMed]
  33. Iqubal, A.; Iqubal, M.K.; Sharma, S.; Ansari, M.A.; Najmi, A.K.; Ali, S.M.; Ali, J.; Haque, S.E. Molecular mechanism involved in cyclophosphamide-induced cardiotoxicity: Old drug with a new vision. Life Sci. 2019, 218, 112–131. [Google Scholar] [CrossRef] [PubMed]
  34. Chaar, M.; Kamta, J.; Ait-Oudhia, S. Mechanisms.s..s, monitoring, and management of tyrosine kinase inhibitors-associated cardiovascular toxicities. Onco Targets Ther. 2018, 11, 6227–6237. [Google Scholar] [CrossRef] [Green Version]
  35. Palaskas, N.; Lopez-Mattei, J.; Durand, J.B.; Iliescu, C.; Deswal, A. Immune Checkpoint Inhibitor Myocarditis: Pathophysiological Characteristics, Diagnosis, and Treatment. J. Am. Heart Assoc. 2020, 9, e013757. [Google Scholar] [CrossRef]
  36. Caspi, O.; Aronson, D. Surviving Cancer without a Broken Heart. Rambam. Maimonides Med. J. 2019, 10, e0012. [Google Scholar] [CrossRef]
  37. Xu, X.; Sutak, R.; Richardson, D.R. Iron chelation by clinically relevant anthracyclines: Alteration in expression of iron-regulated genes and atypical changes in intracellular iron distribution and trafficking. Mol. Pharmacol. 2008, 73, 833–844. [Google Scholar] [CrossRef] [Green Version]
  38. Ichikawa, Y.; Ghanefar, M.; Bayeva, M.; Wu, R.; Khechaduri, A.; Naga Prasad, S.V.; Mutharasan, R.K.; Naik, T.J.; Ardehali, H. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J. Clin. Investig. 2014, 124, 617–630. [Google Scholar] [CrossRef] [Green Version]
  39. Eschenhagen, T.; Force, T.; Ewer, M.S.; de Keulenaer, G.W.; Suter, T.M.; Anker, S.D.; Avkiran, M.; de Azambuja, E.; Balligand, J.L.; Brutsaert, D.L.; et al. Cardiovascular side effects of cancer therapies: A position statement from the Heart Failure Association of the European Society of Cardiology. Eur. J. Heart Fail. 2011, 13, 1–10. [Google Scholar] [CrossRef]
  40. Arinno, A.; Maneechote, C.; Khuanjing, T.; Ongnok, B.; Prathumsap, N.; Chunchai, T.; Arunsak, B.; Kerdphoo, S.; Shinlapawittayatorn, K.; Chattipakorn, S.C.; et al. Cardioprotective effects of melatonin and metformin against doxorubicin-induced cardiotoxicity in rats are through preserving mitochondrial function and dynamics. Biochem. Pharmacol. 2021, 192, 114743. [Google Scholar] [CrossRef]
  41. Gronek, P.; Wielinski, D.; Cyganski, P.; Rynkiewicz, A.; Zajac, A.; Maszczyk, A.; Gronek, J.; Podstawski, R.; Czarny, W.; Balko, S.; et al. A Review of Exercise as Medicine in Cardiovascular Disease: Pathology and Mechanism. Aging Dis. 2020, 11, 327–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Powers, S.K.; Deminice, R.; Ozdemir, M.; Yoshihara, T.; Bomkamp, M.P.; Hyatt, H. Exercise-induced oxidative stress: Friend or foe? J. Sport Health Sci. 2020, 9, 415–425. [Google Scholar] [CrossRef]
  43. Moore, S.C.; Lee, I.M.; Weiderpass, E.; Campbell, P.T.; Sampson, J.N.; Kitahara, C.M.; Keadle, S.K.; Arem, H.; Berrington de Gonzalez, A.; Hartge, P.; et al. Association of Leisure-Time Physical Activity with Risk of 26 Types of Cancer in 1.44 Million Adults. JAMA Intern. Med. 2016, 176, 816–825. [Google Scholar] [CrossRef] [PubMed]
  44. Scott, J.M.; Zabor, E.C.; Schwitzer, E.; Koelwyn, G.J.; Adams, S.C.; Nilsen, T.S.; Moskowitz, C.S.; Matsoukas, K.; Iyengar, N.M.; Dang, C.T.; et al. Efficacy of Exercise Therapy on Cardiorespiratory Fitness in Patients with Cancer: A Systematic Review and Meta-Analysis. J. Clin. Oncol. 2018, 36, 2297–2305. [Google Scholar] [CrossRef] [PubMed]
  45. Jones, L.W.; Hornsby, W.E.; Freedland, S.J.; Lane, A.; West, M.J.; Moul, J.W.; Ferrandino, M.N.; Allen, J.D.; Kenjale, A.A.; Thomas, S.M.; et al. Effects of nonlinear aerobic training on erectile dysfunction and cardiovascular function following radical prostatectomy for clinically localized prostate cancer. Eur. Urol. 2014, 65, 852–855. [Google Scholar] [CrossRef] [Green Version]
  46. Adams, S.C.; DeLorey, D.S.; Davenport, M.H.; Stickland, M.K.; Fairey, A.S.; North, S.; Szczotka, A.; Courneya, K.S. Effects of high-intensity aerobic interval training on cardiovascular disease risk in testicular cancer survivors: A phase 2 randomized controlled trial. Cancer 2017, 123, 4057–4065. [Google Scholar] [CrossRef]
  47. Gilchrist, S.C.; Barac, A.; Ades, P.A.; Alfano, C.M.; Franklin, B.A.; Jones, L.W.; La Gerche, A.; Ligibel, J.A.; Lopez, G.; Madan, K.; et al. Cardio-Oncology Rehabilitation to Manage Cardiovascular Outcomes in Cancer Patients and Survivors: A Scientific Statement From the American Heart Association. Circulation 2019, 139, e997–e1012. [Google Scholar] [CrossRef]
  48. MacVicar, M.G.; Winningham, M.L.; Nickel, J.L. Effects of aerobic interval training on cancer patients’ functional capacity. Nurs. Res. 1989, 38, 348–351. [Google Scholar] [CrossRef]
  49. Segal, R.; Evans, W.; Johnson, D.; Smith, J.; Colletta, S.; Gayton, J.; Woodard, S.; Wells, G.; Reid, R. Structured exercise improves physical functioning in women with stages I and II breast cancer: Results of a randomized controlled trial. J. Clin. Oncol. 2001, 19, 657–665. [Google Scholar] [CrossRef]
  50. van Waart, H.; Stuiver, M.M.; van Harten, W.H.; Geleijn, E.; Kieffer, J.M.; Buffart, L.M.; de Maaker-Berkhof, M.; Boven, E.; Schrama, J.; Geenen, M.M.; et al. Effect of Low-Intensity Physical Activity and Moderate- to High-Intensity Physical Exercise During Adjuvant Chemotherapy on Physical Fitness, Fatigue, and Chemotherapy Completion Rates: Results of the PACES Randomized Clinical Trial. J. Clin. Oncol. 2015, 33, 1918–1927. [Google Scholar] [CrossRef] [Green Version]
  51. Haykowsky, M.J.; Mackey, J.R.; Thompson, R.B.; Jones, L.W.; Paterson, D.I. Adjuvant trastuzumab induces ventricular remodeling despite aerobic exercise training. Clin. Cancer Res. 2009, 15, 4963–4967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Segal, R.J.; Reid, R.D.; Courneya, K.S.; Sigal, R.J.; Kenny, G.P.; Prud’Homme, D.G.; Malone, S.C.; Wells, G.A.; Scott, C.G.; Slovinec D’Angelo, M.E. Randomized controlled trial of resistance or aerobic exercise in men receiving radiation therapy for prostate cancer. J. Clin. Oncol. 2009, 27, 344–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Jones, L.W.; Habel, L.A.; Weltzien, E.; Castillo, A.; Gupta, D.; Kroenke, C.H.; Kwan, M.L.; Quesenberry, C.P., Jr.; Scott, J.; Sternfeld, B.; et al. Exercise and Risk of Cardiovascu.ular Events in Women with Nonmetastatic Breast Cancer. J. Clin. Oncol. 2016, 34, 2743–2749. [Google Scholar] [CrossRef] [Green Version]
  54. Schmid, D.; Leitzmann, M.F. Association between physical activity and mortality among breast cancer and colorectal cancer survivors: A systematic review and meta-analysis. Ann. Oncol. 2014, 25, 1293–1311. [Google Scholar] [CrossRef]
  55. Scott, J.M.; Li, N.; Liu, Q.; Yasui, Y.; Leisenring, W.; Nathan, P.C.; Gibson, T.; Armenian, S.H.; Nilsen, T.S.; Oeffinger, K.C.; et al. Association of Exercise with Mortality in Adult Survivors of Childhood Cancer. JAMA Oncol. 2018, 4, 1352–1358. [Google Scholar] [CrossRef]
  56. Pinto, B.M.; Papandonatos, G.D.; Goldstein, M.G.; Marcus, B.H.; Farrell, N. Home-based physical activity intervention for colorectal cancer survivors. Psychooncology 2013, 22, 54–64. [Google Scholar] [CrossRef] [PubMed]
  57. Courneya, K.S.; Friedenreich, C.M.; Quinney, H.A.; Fields, A.L.; Jones, L.W.; Fairey, A.S. A randomized trial of exercise and quality of life in colorectal cancer survivors. Eur. J. Cancer Care 2003, 12, 347–357. [Google Scholar] [CrossRef]
  58. Zhou, Y.; Zhu, J.; Gu, Z.; Yin, X. Efficacy of Exercise Interventions in Patients with Acute Leukemia: A Meta-Analysis. PLoS ONE 2016, 11, e0159966. [Google Scholar] [CrossRef]
  59. Courneya, K.S.; Sellar, C.M.; Stevinson, C.; McNeeely, M.L.; Peddle, C.J.; Friedenreich, C.M.; Tankel, K.; Basi, S.; Chua, N.; Mazurek, A.; et al. Randomized controlled trial of the effects of aerobic exercise on physical functioning and quality of life in lymphoma patients. J. Clin. Oncol. 2009, 27, 4605–4612. [Google Scholar] [CrossRef] [Green Version]
  60. Speck, R.M.; Courneya, K.S.; Masse, L.C.; Duval, S.; Schmitz, K.H. An update of controlled physical activity trials in cancer survivors: A systematic review and meta-analysis. J. Cancer Surviv. 2010, 4, 87–100. [Google Scholar] [CrossRef]
  61. Jones, L.W.; Liang, Y.; Pituskin, E.N.; Battaglini, C.L.; Scott, J.M.; Hornsby, W.E.; Haykowsky, M. Effect of exercise training on peak oxygen consumption in patients with cancer: A meta-analysis. Oncologist 2011, 16, 112–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Courneya, K.S.; Segal, R.J.; Mackey, J.R.; Gelmon, K.; Reid, R.D.; Friedenreich, C.M.; Ladha, A.B.; Proulx, C.; Vallance, J.K.; Lane, K.; et al. Effects of aerobic and resistance exercise in breast cancer patients receiving adjuvant chemotherapy: A multicenter randomized controlled trial. J. Clin. Oncol. 2007, 25, 4396–4404. [Google Scholar] [CrossRef] [PubMed]
  63. Pelliccia, A.; Sharma, S.; Gati, S.; Back, M.; Borjesson, M.; Caselli, S.; Collet, J.P.; Corrado, D.; Drezner, J.A.; Halle, M.; et al. 2020 ESC Guidelines on Sports Cardiology and Exercise in Patients with Cardiovascular Disease. Rev. Esp. Cardiol. 2021, 74, 545. [Google Scholar] [CrossRef]
  64. Flynn, K.E.; Pina, I.L.; Whellan, D.J.; Lin, L.; Blumenthal, J.A.; Ellis, S.J.; Fine, L.J.; Howlett, J.G.; Keteyian, S.J.; Kitzman, D.W.; et al. Effects of exercise training on health status in patients with chronic heart failure: HF-ACTION randomized controlled trial. JAMA 2009, 301, 1451–1459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. O’Connor, C.M.; Whellan, D.J.; Lee, K.L.; Keteyian, S.J.; Cooper, L.S.; Ellis, S.J.; Leifer, E.S.; Kraus, W.E.; Kitzman, D.W.; Blumenthal, J.A.; et al. Efficacy and safety of exercise training in patients with chronic heart failure: HF-ACTION randomized controlled trial. JAMA 2009, 301, 1439–1450. [Google Scholar] [CrossRef]
  66. Taylor, R.S.; Walker, S.; Smart, N.A.; Piepoli, M.F.; Warren, F.C.; Ciani, O.; O’Connor, C.; Whellan, D.; Keteyian, S.J.; Coats, A.; et al. Impact of exercise-based cardiac rehabilitation in patients with heart failure (ExTraMATCH II) on mortality and hospitalisation: An individual patient data meta-analysis of randomised trials. Eur. J. Heart Fail. 2018, 20, 1735–1743. [Google Scholar] [CrossRef] [Green Version]
  67. Taylor, R.S.; Long, L.; Mordi, I.R.; Madsen, M.T.; Davies, E.J.; Dalal, H.; Rees, K.; Singh, S.J.; Gluud, C.; Zwisler, A.D. Exercise-Based Rehabilitation for Heart Failure: Cochrane Systematic Review, Meta-Analysis, and Trial Sequential Analysis. JACC Heart Fail. 2019, 7, 691–705. [Google Scholar] [CrossRef]
  68. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Bohm, M.; Burri, H.; Butler, J.; Celutkiene, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: Developed by the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) with the special contribution of the Heart Failure Association (HFA) of the ESC. Rev. Esp. Cardiol. 2022, 75, 523. [Google Scholar] [CrossRef]
  69. Pandey, A.; Parashar, A.; Kumbhani, D.; Agarwal, S.; Garg, J.; Kitzman, D.; Levine, B.; Drazner, M.; Berry, J. Exercise training in patients with heart failure and preserved ejection fraction: Meta-analysis of randomized control trials. Circ. Heart Fail. 2015, 8, 33–40. [Google Scholar] [CrossRef] [Green Version]
  70. Kitzman, D.W.; Brubaker, P.; Morgan, T.; Haykowsky, M.; Hundley, G.; Kraus, W.E.; Eggebeen, J.; Nicklas, B.J. Effect of Caloric Restriction or Aerobic Exercise Training on Peak Oxygen Consumption and Quality of Life in Obese Older Patients with Heart Failure with Preserved Ejection Fraction: A Randomized Clinical Trial. JAMA 2016, 315, 36–46. [Google Scholar] [CrossRef]
  71. Kitagawa, T.; Hidaka, T.; Watanabe, N.; Naka, M.; Yamaguchi, M.; Kanai, K.; Isobe, M.; Kihara, Y.; Nakano, Y.; Investigators, R.-H. Current conditions and significance of outpatient cardiac rehabilitation and home nursing-care services in heart failure patients with mid-range or preserved ejection fraction: Post-hoc analysis of the REAL-HF registry. Heart Vessel. 2022, 37, 745–754. [Google Scholar] [CrossRef] [PubMed]
  72. Beatty, A.L.; Truong, M.; Schopfer, D.W.; Shen, H.; Bachmann, J.M.; Whooley, M.A. Geographic Variation in Cardiac Rehabilitation Participation in Medicare and Veterans Affairs Populations: Opportunity for Improvement. Circulation 2018, 137, 1899–1908. [Google Scholar] [CrossRef] [PubMed]
  73. Pandey, A.; Keshvani, N.; Zhong, L.; Mentz, R.J.; Pina, I.L.; DeVore, A.D.; Yancy, C.; Kitzman, D.W.; Fonarow, G.C. Temporal Trends and Factors Associated with Cardiac Rehabilitation Participation Among Medicare Beneficiaries with Heart Failure. JACC Heart Fail. 2021, 9, 471–481. [Google Scholar] [CrossRef] [PubMed]
  74. Thomas, R.J.; Beatty, A.L.; Beckie, T.M.; Brewer, L.C.; Brown, T.M.; Forman, D.E.; Franklin, B.A.; Keteyian, S.J.; Kitzman, D.W.; Regensteiner, J.G.; et al. Home-Based Cardiac Rehabilitation: A Scientific Statement From the American Association of Cardiovascular and Pulmonary Rehabilitation, the American Heart Association, and the American College of Cardiology. J. Am. Coll. Cardiol. 2019, 74, 133–153. [Google Scholar] [CrossRef]
  75. Kirkham, A.A.; Virani, S.A.; Bland, K.A.; McKenzie, D.C.; Gelmon, K.A.; Warburton, D.E.R.; Campbell, K.L. Exercise training affects hemodynamics not cardiac function during anthracycline-based chemotherapy. Breast Cancer Res. Treat. 2020, 184, 75–85. [Google Scholar] [CrossRef]
  76. Howden, E.J.; Bigaran, A.; Beaudry, R.; Fraser, S.; Selig, S.; Foulkes, S.; Antill, Y.; Nightingale, S.; Loi, S.; Haykowsky, M.J.; et al. Exercise as a diagnostic and therapeutic tool for the prevention of cardiovascular dysfunction in breast cancer patients. Eur. J. Prev. Cardiol. 2019, 26, 305–315. [Google Scholar] [CrossRef]
  77. Kirkham, A.A.; Shave, R.E.; Bland, K.A.; Bovard, J.M.; Eves, N.D.; Gelmon, K.A.; McKenzie, D.C.; Virani, S.A.; Stohr, E.J.; Warburton, D.E.R.; et al. Protective effects of acute exercise prior to doxorubicin on cardiac function of breast cancer patients: A proof-of-concept RCT. Int. J. Cardiol. 2017, 245, 263–270. [Google Scholar] [CrossRef]
  78. Jones, L.W.; Douglas, P.S.; Khouri, M.G.; Mackey, J.R.; Wojdyla, D.; Kraus, W.E.; Whellan, D.J.; O’Connor, C.M. Safety and efficacy of aerobic training in patients with cancer who have heart failure: An analysis of the HF-ACTION randomized trial. J. Clin. Oncol. 2014, 32, 2496–2502. [Google Scholar] [CrossRef] [Green Version]
  79. Abraham, W.T.; Adamson, P.B.; Bourge, R.C.; Aaron, M.F.; Costanzo, M.R.; Stevenson, L.W.; Strickland, W.; Neelagaru, S.; Raval, N.; Krueger, S.; et al. Wireless pulmonary artery haemodynamic monitoring in chronic heart failure: A randomised controlled trial. Lancet 2011, 377, 658–666. [Google Scholar] [CrossRef]
  80. Perl, L.; Meerkin, D.; D’Amario, D.; Avraham, B.B.; Gal, T.B.; Weitsman, T.; Hasin, T.; Ince, H.; Feickert, S.; D’Ancona, G.; et al. The V-LAP System for Remote Left Atrial Pressure Monitoring of Patients with Heart Failure: Remote Left Atrial Pressure Monitoring. J. Card. Fail. 2022, 28, 963–972. [Google Scholar] [CrossRef]
  81. Wyatt, K.D. A Smartwatch to Identify Atrial Fibrillation. N. Engl. J. Med. 2020, 382, 975. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Common mechanisms mediating cardiotoxicity and positively affected by exercise. DM: Diabetes Mellitus.
Figure 1. Common mechanisms mediating cardiotoxicity and positively affected by exercise. DM: Diabetes Mellitus.
Life 12 01006 g001
Table 1. Mechanisms of cardiotoxicity of common cancer therapeutics.
Table 1. Mechanisms of cardiotoxicity of common cancer therapeutics.
Drug Name/ClassMechanism of CardiotoxicityReferences
AnthracyclinesGeneration of excess free radicals[17,18,19]
Accumulation of iron in the mitochondria[37,38]
Top2β inhibition-breakage of DNA and induction of programmed cardiomyocyte death[20]
Dysregulation of cardiomyocyte autophagy[21]
TrastuzumabActivation of autophagy-inhibitory Erk/mTOR/Ulk1 signaling cascade[25]
Interruption of ErbB4/ErbB2 heterodimerization[23,24]
CyclophosphamideDepletion of antioxidants/ATP level[33]
Endothelial damage[33]
Enhanced pro-inflammatory/pro-apoptotic activities[33]
CisplatinInduction of nuclear and mitochondrial DNA damage[31]
Oxidative stress and mitochondrial dysfunction[31,32]
5-FluorouracilCoronary vasospasm through Protein kinase C and Endothelin-I[26,27]
Direct myocardial injury: Alpha-fluoro-beta-alanine (AFBA)[28]
Vascular endothelial dysfunction: direct toxic effect and reactive oxygen species production[29]
Tyrosine kinase inhibitorsOxidative stress, mitochondrial damage, and apoptosis of cardiomyocytes[34]
Immune checkpoint inhibitorsImmune inflammation with T-cell infiltration of the myocardium[30,35]
Oxidative stress[30,35]
Table 2. Summary of studies evaluating exercise rehabilitation for cancer patients.
Table 2. Summary of studies evaluating exercise rehabilitation for cancer patients.
Author/YearPatients CharacteristicOutcomesNum of PatientsExercise TypeResults
Scott, J.M. et al., 2018 [44]mixed (meta analysis) cancer patientsCRF3632aerobic and combined aerobic and resistance therapyincrease in VO2peak
MacVicar, M.G. et al., 1989 [48]breast cancerCRF45aerobic interval trainingsignificant increase in VO2Lmax
Segal, R. et al., 2001 [49]breast cancerCRF123self-directed or supervised exerciseincreased in Short Form-36 physical functioning scale
Van Waart, H. et al., 2015 [50]breast cancerCRF230home-based and supervised exercise programsprevention of decline in CRF, increase physical functioning
Haykowsky, M.J. et al., 2009 [51]trastuzumab-treated breast cancerCRF + LV remodeling17aerobic trainingincreased CRF, did not prevent LV remodeling
Segal et al., 2009 [52]radiation treated prostate cancerCRF, fatigue121resistance or aerobic trainingincreased CRF, decreased fatigue
Jones, L.W. et al., 2016 [53]breast cancerCVE2973leisure-time recreational exercisedecrease in CVE
Schmid, D. et al., 2014 [54]breast and colorectal cancer (meta analysis)mortality49,095physical activitydecreased mortality
Jones, L.W. et al., 2014 [45]post-surgical prostate cancerCRF50Aerobic Trainingincreased CRF
Scott, J.M. et al., 2018 [55]adult survivors of childhood cancermortality15,450questionnaire-completed study on physical activity levelsdecrease in all-cause mortality
Adams, S.C. et al., 2017 [46]testicular cancerCRF63aerobic interval trainingincrease in VO2peak
Pinto, B.M. et al., 2013 [56]colorectal cancerCRF46telephone counseling to support exerciseincrease in exercise duration and fitness
Courneya, K.S. et al., 2003 [57]colorectal cancerquality of life102home-based exercise interventionno change in outcome
Zhou, Y. et al., 2016 [58]acute leukemia (meta-analysis)CRF314exerciseincreased CRF
Courneya, K.S. et al., 2009 [59]lymphomaCRF122aerobic exercise trainingincreased CRF
Speck, R.M. et al., 2010 [60]mixed (meta analysis) cancer patientsCRF6838physical activityincreased CRF
Jones, L.W. et al., 2011 [61]mixed (meta analysis) cancer patientsCRF571supervised exercise trainingincrease in VO2peak
Courneya, K.S. et al., 2007 [62]breast cancerCRF + quality of life242aerobic and resistance exerciseincreased CRF, no change in quality of life
Table 3. Cardiac exercise rehabilitation effects on cardiovascular aspects of cancer patients during therapy with cardiotoxic drugs. Systemic vascular resistance: SVR; ejection fraction: EF; left ventricle: LV; B-type natriuretic peptide: BNP; N-terminal prohormone of brain natriuretic peptide: NT-proBNP.
Table 3. Cardiac exercise rehabilitation effects on cardiovascular aspects of cancer patients during therapy with cardiotoxic drugs. Systemic vascular resistance: SVR; ejection fraction: EF; left ventricle: LV; B-type natriuretic peptide: BNP; N-terminal prohormone of brain natriuretic peptide: NT-proBNP.
HemodynamicsSVR [75] Life 12 01006 i001
Cardiac output [76] Life 12 01006 i002
Blood Pressure [75] Life 12 01006 i002
EchocardiographicEF [75,76,77] Life 12 01006 i002 Life 12 01006 i003
LV remodeling [75] Life 12 01006 i002
LV longitudinal strain [75,76] Life 12 01006 i002
E/A ratio [75] Life 12 01006 i002
LV mass [75] Life 12 01006 i002 Life 12 01006 i003
BiomarkersTroponin [76] Life 12 01006 i002
NT-proBNP [77] Life 12 01006 i001
Cardiorespiratory functionVO2peak [76] Life 12 01006 i003
Muscle strength [50] Life 12 01006 i003
Fatigue [50] Life 12 01006 i001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Elad, B.; Habib, M.; Caspi, O. Cardio-Oncology Rehabilitation—Present and Future Perspectives. Life 2022, 12, 1006. https://doi.org/10.3390/life12071006

AMA Style

Elad B, Habib M, Caspi O. Cardio-Oncology Rehabilitation—Present and Future Perspectives. Life. 2022; 12(7):1006. https://doi.org/10.3390/life12071006

Chicago/Turabian Style

Elad, Boaz, Manhal Habib, and Oren Caspi. 2022. "Cardio-Oncology Rehabilitation—Present and Future Perspectives" Life 12, no. 7: 1006. https://doi.org/10.3390/life12071006

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop