Next Article in Journal
Using Extracellular Vesicles Released by GDNF-Transfected Macrophages for Therapy of Parkinson Disease
Next Article in Special Issue
Understanding Metabolic Alterations in Cancer Cachexia through the Lens of Exercise Physiology
Previous Article in Journal
Examples of Inverse Comorbidity between Cancer and Neurodegenerative Diseases: A Possible Role for Noncoding RNA
Previous Article in Special Issue
Depleting Ly6G Positive Myeloid Cells Reduces Pancreatic Cancer-Induced Skeletal Muscle Atrophy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiac Remodeling in Cancer-Induced Cachexia: Functional, Structural, and Metabolic Contributors

by
Michael P. Wiggs
1,
Anna G. Beaudry
1 and
Michelle L. Law
2,*,†
1
Department of Health, Human Performance, and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA
2
Department of Human Sciences and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA
*
Author to whom correspondence should be addressed.
Mailing address: Department of Human Sciences and Design, Robbins College of Health and Human Sciences, Baylor University, One Bear Place #97346, Waco, TX 76798, USA.
Cells 2022, 11(12), 1931; https://doi.org/10.3390/cells11121931
Submission received: 29 April 2022 / Revised: 1 June 2022 / Accepted: 8 June 2022 / Published: 15 June 2022
(This article belongs to the Special Issue Cancer-Induced Cachexia)

Abstract

:
Cancer cachexia is a syndrome of progressive weight loss and muscle wasting occurring in many advanced cancer patients. Cachexia significantly impairs quality of life and increases mortality. Cardiac atrophy and dysfunction have been observed in patients with cachexia, which may contribute to cachexia pathophysiology. However, relative to skeletal muscle, little research has been carried out to understand the mechanisms of cardiomyopathy in cachexia. Here, we review what is known clinically about the cardiac changes occurring in cachexia, followed by further discussion of underlying physiological and molecular mechanisms contributing to cachexia-induced cardiomyopathy. Impaired cardiac contractility and relaxation may be explained by a complex interplay of significant heart muscle atrophy and metabolic remodeling, including mitochondrial dysfunction. Because cardiac muscle has fundamental differences compared to skeletal muscle, understanding cardiac-specific effects of cachexia may bring light to unique therapeutic targets and ultimately improve clinical management for patients with cancer cachexia.

1. Introduction

1.1. Cancer Cachexia Overview

Cancer cachexia is a multifactorial syndrome of progressive and unintentional weight loss. It occurs in 30% of all cancer patients and 70–90% of patients with cancers of the lung, liver, and gastrointestinal tract [1]. This condition is thought to contribute to ~20% of cancer deaths [2]. Prevalence statistics for cancer cachexia are not included in the national cancer records of any country [3], so these suggested percentages may underestimate the true prevalence. Cancer is a leading cause of death worldwide, accounting for nearly 10 million deaths in 2020 [4]. Many of these cancer deaths result from cancers commonly associated with cachexia, such as gastric, pulmonary, pancreatic, esophageal, hepatic, and colorectal cancers [5].
An international Delphi consensus definition and diagnostic classification for cancer cachexia was published in 2011 [6]. As outlined in this document, a cancer cachexia diagnosis should be made when body weight loss is greater than 5% within six months, when body mass index (BMI) is less than 20 kg/m2 in combination with body weight loss of greater than 2%, or when appendicular skeletal muscle index is consistent with sarcopenia and weight loss is greater than 2% [6]. These are the current diagnostic criteria; however, uncertainty remains as to whether more specificity may be beneficial for purposes of identification and prevention. Specifically, body composition (fat vs. lean mass) and changes in body composition over time may be important predictors of morbidity and mortality [7,8]. The discovery of specific biomarkers that predict for cancer-induced weight loss would also be diagnostically advantageous [9].
In this multifactorial syndrome, there is ongoing muscle and adipose wasting, leading to progressive functional decline. Patients often exhibit reduced tolerance and effectiveness of cancer therapies [10], such as radiotherapy and chemotherapy, as well as increased risk of chemotoxicity. In addition to increased mortality, cancer cachexia causes profound fatigue and weakness, decreases quality of life [11], increases the occurrence of cancer-related complications [12], and increases medical costs [13]. Although anamorelin–a ghrelin receptor agonist–was recently approved to treat cachexia in Japan [14], therapeutic options remain very limited. Furthermore, because of the complexity of cachexia pathology, a multimodal treatment strategy is likely required to adequately address the multiple facets of this syndrome [15].
Cancer cachexia can be described as a condition involving systemic inflammation, metabolic dysregulation, and decreased food intake leading to energy imbalance [3]. Unlike starvation, cancer cachexia cannot be completely reversed by adequate nutrition alone [16,17]. Resting energy expenditure is often elevated—at least in part due to tumor metabolism [18,19,20], inflammation [21,22,23], mitochondrial dysfunction [24], and futile cycling [25,26,27]—thus, promoting a negative energy balance [28,29]. Abnormalities in protein synthesis and degradation, as well as amino acid transport, are observed in cachectic skeletal muscle. Proteolysis is highly upregulated while the rate of protein synthesis declines [30,31]. Muscle breakdown during cancer cachexia occurs with significant involvement of the ubiquitin proteasome system (UPS) and autophagy lysosome system (ALS) [32]. There is also evidence of increased apoptosis, impaired muscle regenerative capacity [33], and increased branched-chain amino acid oxidation [30]. In combination, these alterations lead to the highly catabolic state that is characteristic of cancer-induced cachexia. Evidence suggests that other tissues and organs, including heart, adipose, brain, liver, bone, blood, and gut, are also adversely affected by cachexia and may contribute to cachexia pathology [30,34]. These systemic implications are thought to be caused by inflammatory factors released from both tumor and host tissues.

1.2. Cancer Cachexia and the Heart

In addition to cancer cachexia’s significant deleterious effects on skeletal muscle size and function, increasing evidence points to adverse effects of cachexia on cardiac muscle as well. However, relative to skeletal muscle, little work has been done clinically to identify effects of cancer cachexia on the heart or experimentally to uncover underlying mechanisms driving cachexia-induced cardiac functional changes. Nevertheless, the importance of understanding cardiac pathology in cancer cachexia is emphasized by the fact that cachexia is also present in a high percentage of heart failure patients [35], uncovering a potential link between cardiac disease and skeletal muscle wasting. Due to many similar mediating factors between cancer cachexia and heart failure, it is plausible that cancer-induced cachexia may contribute to cardiac dysfunction, and cardiac dysfunction may in turn exacerbate cachexia to create a vicious cycle contributing to morbidity and mortality [36] (Figure 1).
The increasing emphasis in recent years on evaluating and treating heart disease in cancer patients has been driven in large part by the known cardiotoxicity of many chemotherapeutic agents as well as pre-existing cardiovascular disease in many cancer patients [37,38]. These factors have led to growth in the field of cardio-oncology [39]. Furthermore, improvements in early cancer detection and development of more targeted and effective cancer therapies are leading to remission or stabilized disease with increased length of life, even for some patients with advanced disease. As a result, more cancer patients die of causes not directly related to tumor burden. According to the National Cancer Institute’s Surveillance, Epidemiology, and End Results (SEER) data, deaths from cancer among patients who have had a cancer diagnosis have progressively declined from approximately 80% to 60% over a 40-year period from 1973 to 2012, and deaths from other causes have increased [40]. Heart disease is the greatest cause of non-cancer death among cancer patients, accounting for approximately half of all non-cancer deaths [40]. The risk of heart-related death is 2-fold higher in patients who have had a cancer diagnosis compared to the general population [40]. Among patients actively being treated for cancer, two different studies found that ~7% (35/506 patients) [41] and 9% (75/816 patients) [42] died of cardiac failure or acute myocardial infarction and not directly from the tumor.
To date, the field of cardio-oncology has not focused largely on cachexia as a contributor to cardiac disease in cancer patients. Indeed, teasing apart individual effects of cancer therapies, pre-existing heart disease, and cachexia in a clinical setting is difficult in many situations. However, both clinical and pre-clinical evidence of direct effects of cachexia on the heart is growing. The purpose of this review is to provide a discussion of what has been observed clinically and what is known mechanistically related to the effects of cachexia on cardiac (1) functional remodeling, (2) structural remodeling, and (3) metabolic remodeling, and what questions remain. An understanding of cancer cachexia-induced cardiac pathophysiology is imperative to developing optimal treatment plans and therapeutic strategies for cancer patients. As cardiac muscle has minimal regenerative capacity [43], protecting the heart is essential to increasing both quality and length of life for individuals with cancer diagnoses.

2. Functional Remodeling

Evidence of impaired cardiac function in cancer-induced cachexia has grown in recent years. Importantly, cardiac functional deficits may directly contribute to cachexia-related fatigue, consequently diminishing whole body functional capacity and quality of life. The following section reviews what is known about cardiac function in patients with cachexia and follows with physiological underpinnings in pre-clinical models.

2.1. Clinical Observations of Cardiac Functional Changes

Elevated heart rate has been observed in multiple clinical studies of cancer patients, both with and without overt cachexia. Heart rate elevation was present across multiple types of cancer, including breast [44], colon [45,46], lung [45], and pancreatic [45], in both treatment-naïve [45,46] and chemotherapy treated groups [45]. Importantly, elevated heart rate was associated with increased mortality [44,45]. Increased sympathetic nervous system (SNS) activation is a likely contributor to increased chronotropy [47]. Decreased heart rate variability [46] and increased serum B-type natriuretic peptide (BNP) [48] also provide evidence of impaired sympathoadrenal signaling in cachectic patients. Impaired oxygen uptake (VO2 max) [46] and anemia [49] may have a role in SNS activation in cachexia. Moreover, increased ergoreflex activation due to metabolic and structural changes in skeletal muscle has also been implicated in increased SNS activity [50,51], although the specific effects of ergoreflex activation in cancer-induced cachexia have not been studied to our knowledge.
It was hypothesized that cachexia may increase susceptibility to electrical abnormalities in the hearts of cachectic patients as a result of cardiac atrophy and increased ventricular wall stress [52]. Electrocardiogram (ECG) measurements, including PQ, QRS, and QTc duration, were not different between cancer patients and controls when data were collected at a single time point [45]. However, when a subset of these patients underwent 24-hour ECG testing, 9/120 cancer patients and 0/43 control patients had ≥1 episodes of non-sustained ventricular tachycardia, which predicted for increased mortality after 5 years [53]. Although this study did not specifically look at cachexia, colorectal, lung, and pancreatic cancer patients included in this cohort have a high incidence of cancer-induced weight loss [1].
Cardiac contractile function in cancer patients has also been measured by echocardiography studies. Treatment-naïve colorectal cancer (CRC) patients had decreased left ventricular ejection fraction (LVEF)–a measure of systolic function–compared to control subjects at a single time point [46]. Stage IV non-small cell lung cancer (NSCLC) patients underwent echocardiography before and ~4 months following carboplatin therapy. LVEF and global longitudinal strain (GLS), another measure of systolic function, were significantly decreased at 4 months compared to baseline [54]. The effects of carboplatin versus cancer on cardiac function cannot be determined from this study. Carboplatin is in the class of drugs known as alkylating agents, some of which may have cardiotoxic effects [55]. However, loss of left ventricular mass was associated with the largest decrease in GLS [54], suggesting an association between functional decline and cardiac atrophy. It is important to note, despite significant differences in LVEF in the former two studies, neither study reported mean LVEF values that would be considered clinically abnormal (< 50%). A study in treatment-naïve NSCLC patients without a history of cardiovascular disease did identify 7/70 patients with LVEF < 50%. Of these seven patients, five were identified as cachectic and one was identified as pre-cachectic [56]. Because cardiac dysfunction is also a risk factor for the development of cachexia [35], the causal direction between cachexia and systolic dysfunction is unknown. However, this study identifies a significant association between cachexia and cardiac abnormalities, which complicates the clinical picture for this subset of cancer patients. A summary of clinical studies identifying functional cardiac remodeling is found in Table 1.

2.2. Mechanistic Contributors to Cardiac Functional Changes

Echocardiographic abnormalities have also been observed in multiple pre-clinical models of cachexia, providing clinical relevance to these models. Colon-26 Adenocarcinoma (C26) [61,62,63,64,65,66,67,68,69], Yoshida AH-130 Ascites [48,70,71], and Ehrlich Ascites Carcinoma [72] models all show diminished in vivo cardiac performance. Cardiac function in the Lewis Lung Carcinoma (LLC) model appears to be sex-specific, with males having impaired function and females showing protection against tumor-induced functional changes [69,73]. Most studies identify systolic functional deficits measured by decreased fractional shortening (FS) or percent ejection fraction (%EF) [48,61,62,64,65,67,68,69,70,71,72], although stroke volume [62,71] and cardiac output [48,70] are also reported. Fewer studies have assessed in vivo diastolic performance. Xu et al. reported no difference in the E/A ratio [65]; E/A is the ratio of early to late ventricle filling. Diastolic dysfunction is characterized by a non-compliant heart, leading to less early filling and a lower E/A ratio. Tei index, a measure of global systolic and diastolic left ventricle function, was unchanged in one study [65] and increased (indicating worsened function) in another [68]. Despite little difference in diastolic function in echocardiography studies, invasive in vivo hemodynamics measurements identified significantly impaired diastolic function–measured by left ventricular end-diastolic pressure (LVEDP) and negative pressure derivative (-dP/dt; relaxation rate) in the Yoshida AH-130 rat cachexia model [48]. The increased resting heart rate that has been noted in clinical studies of cachectic patients has not been recapitulated in animal models. Heart rate in pre-clinical echocardiography studies has either been unchanged [64,65,73] or decreased [48,67]. However, telemetry studies may provide a more accurate understanding of heart rate fluctuations and heart rate variability in animal models as echocardiography in rodents must be carried out under anesthesia.
Underlying physiological contributors to impaired in vivo function have been examined in ex vivo isolated hearts to identify functional aberrations outside of the diseased animal [74]. In contrast to in vivo studies, working heart preparations from tumor-bearing cachectic rats unexpectedly had increased baseline function measured by coronary flow, cardiac output, and oxygen consumption [75,76]. Hearts from tumor-bearing rats also had increased β-adrenergic sensitivity, evidenced by a greater increase in heart rate, peak systolic pressure, and positive pressure derivative (+dP/dt; contraction rate) in response to isoproterenol treatment, compared to control hearts [75,76]. In a similar experiment using the C26 mouse model of cachexia, Law and Metzger found Langendorff-perfused hearts had decreased left ventricular developed pressure (LVDP; peak pressure) and +dP/dt, both indicating decreased systolic function [74]. Furthermore, significant impairment in diastolic function was uncovered. Cachectic hearts had diminished -dP/dt and increased LVEDP under pacing stress conditions, indicating incomplete relaxation at higher heart rates [74]. The reason for the discrepancy in findings between Drott et al. [75,76] and Law and Metzger [74] are unclear, although model-specific differences may exist. Additionally, there are differences between the Langendorff and working heart preparations. Langendorff involves retrograde perfusion through the aorta, with a water-filled balloon inserted into the left-ventricle to measure pressure. The heart is paced via electrical stimulation and is thus controlled. Working heart preparations mimic in vivo blood flow, with perfusate entering the left atrium via the pulmonary vein. Hearts are not paced, and the heart pumps perfusate out of the aorta against a resistance [77]. Regardless of the differences in outcome, these experiments are significant as they establish cardiac-intrinsic functional changes between control and tumor-bearing animals under controlled experimental conditions, providing hearts with equal oxygenation and energy substrate availability. More work is needed to identify the mechanistic underpinnings of the differential results between different models and heart preparations.
Changes in isolated cardiac myocyte function have also been uncovered in the C26 model of cachexia, although, similar to ex vivo heart function, findings between studies are somewhat inconsistent. Law and Metzger [74] found a significantly decreased peak height of myocyte contraction, and a slowed rate of contraction and relaxation. Underlying these functional deficits was a profound decrease in calcium cycling. Decreased peak calcium and increased time to peak calcium and 50% calcium decay closely mirrored the deficits in contraction and relaxation, uncovering an important cellular-intrinsic physiological mechanism of impaired contractility and relaxation. Of note, three other studies using the C26 model found decreased in vivo contractility (%EF and FS), but this did not correlate with changes in the peak height of contraction in isolated myocytes. Furthermore, the studies had mixed results regarding myocyte relaxation and changes in calcium cycling [61,62,65]. Two major differences exist between these studies. The former study [74] used male mice that had undergone significant body wasting (~20% difference in body weight between tumor and control groups) and cardiac atrophy (14% difference between tumor and control groups). Female mice with less severe body wasting and no difference in cardiac mass were used in the latter studies [61,62,65]. Importantly, the differences between these studies suggest a role for cachexia severity in cardiac functional deficits, and also reveals potential sex differences between the onset and progression of cardiac dysfunction in cancer-induced cachexia. The time course of cardiomyopathy development and sex differences in cardiac dysfunction in cachexia deserve further investigation.
In conclusion, significant evidence points to in vivo contractility deficits in experimental cachexia. The isolated heart and cardiac myocyte experiments discussed here provide insight into the physiological underpinnings of in vivo cardiac dysfunction. Although findings differ based on the model, functional assay, sex, and cachexia severity, it is evident that heart- and myocyte-intrinsic alterations in cardiac function exist outside of the diseased animal. The molecular mechanisms of these physiological changes are likely complex and multifactorial. Changes in contractility and calcium cycling in cardiac myocytes likely stem from tumor-induced systemic changes including inflammation, altered substrate availability, and changes in sympathoadrenal signaling. These extrinsic signals lead to intrinsic changes in gene and protein expression and post-translational modifications of proteins within the myocyte, converging to induce structural and metabolic remodeling which ultimately affects cardiac performance. Structural and metabolic changes in the heart during cachexia are discussed in the subsequent sections.

3. Structural Remodeling

Numerous organ- and cellular-level structural changes occur in the heart muscle during cachexia development, which may contribute to impaired cardiac function. Most notably, profound atrophy of the heart muscle is found in humans and animal models of cachexia. Underlying the atrophic phenotype are cellular-level changes in sarcomere protein expression and rates of protein synthesis and degradation.

3.1. Clinical Observations of Cardiac Structural Changes

In 1968, Burch et al. described a condition termed the “cachectic heart” that occurred in patients with malignancy and other chronic conditions resulting in malnutrition or inactivity. The cachectic heart was defined as “an acquired pathologic decrease in size, mass, and [epicardial] fat content of the heart with little or no atherosclerosis of the aorta and coronary arteries” [78]. Other observable anatomical and structural changes included lymphocyte infiltration, a flabby appearance of the ventricles, spaces between atrophied muscle fibers, absence of ischemic disease, and an estimated loss of cardiac mass approximately proportional to the total loss of body mass [78]. These morphological changes are in stark contrast to “classic” heart failure, which is characterized by a compensatory hypertrophic remodeling phase, followed by pump failure with dilation and thinning of the ventricular walls [79]. More recent studies have confirmed Burch’s findings of a profound decrease in cardiac mass in cancer patients. In general, an increase in total body weight loss is associated with increased atrophy of the cardiac muscle. Examination of post-mortem cardiac mass in patients with cancer cachexia, cancer patients without cachexia, and patients dying from other causes revealed significantly reduced total cardiac mass and left-ventricle wall thickness with increased fibrosis in cachectic patients compared to both non-cachectic cancer patients and control patients [48]. A retrospective study of 177 patients who died of lung, gastrointestinal, and pancreatic cancer and without history of cardiovascular disease confirmed these findings, also identifying significantly decreased cardiac mass in cachectic patients [57]. However, cachectic patients in this study also had higher rates of chemotherapy and radiotherapy compared to non-cachectic patients, making cachexia-specific effects on cardiac size difficult to ascertain. Nevertheless, heart mass was correlated to body weight in all patients, suggesting body weight loss alone may have some effect on cardiac atrophy [57].
Cardiac atrophy in cancer patients is associated with worsened clinical outcomes. In treatment-naïve advanced pancreatic cancer patients, a decreased left ventricular muscle area (LVMA) predicted shortened overall survival, and left ventricular muscle radiation attenuation (LVMRA) predicted a reduction in both overall and progression-free survival [58]. Muscle radiation attenuation measures muscle density via computed tomography (CT) scan, with lower attenuation scores indicating intramuscular fat deposition. In skeletal muscle, decreased muscle attenuation is associated with decreased strength, independent of total muscle mass [80]. A potential limitation of this study is the relatively infrequent use of CT scans to measure cardiac mass, but an echocardiography study by Kazemi-Bajestani et al. is in agreement with these findings. Kazemi-Bajestani et al. conducted a prospective, longitudinal study of 50 NSCLC patients. Cardiac tissue atrophy over time, measured by echocardiography, was a predictor of disease severity and clinical outcomes [54]. This study included cardiac tissue mass along with skeletal muscle and adipose mass in the analyses. As the number of tissues exhibiting atrophy increased, there was a decrease in physical functioning, chemotherapy response, and survival. Moreover, cardiac atrophy was independently associated with dose-limiting toxicity of chemotherapy [54]. A summary of clinical studies identifying structural remodeling is found in Table 1.

3.2. Mechanistic Contributors to Cardiac Structural Changes

Cardiac atrophy is also noted in multiple pre-clinical models of cancer cachexia [48,67,81], providing consistency between findings in animals and humans with cachexia. Decreased ventricular mass in animals has been attributed to decreased thickness of the interventricular septum [66] and left ventricle posterior wall [66,71] without an increase in left ventricular diameter at diastole [66,71]. These findings, together with decreased cardiac myocyte width in the absence of increased length [74], suggest an atrophic rather than a dilated phenotype, consistent with clinical observations. Underlying cardiac atrophy is an alteration in the expression of myofilament proteins, specifically a decreased total protein expression of myosin heavy chain (MHC) [48,66] and troponin I (cTnI) [66] in whole heart homogenates. A proteomics study in hearts of C26 tumor-bearing mice found an increase in multiple sarcomeric proteins in the soluble protein fraction, including multiple isoforms of MHC, actin, tropomyosin, and troponin, as well as titin. The authors suggested the increase was due to a destabilized sarcomere and increased release of sarcomeric proteins for degradation [82]. Indeed, sarcomeric ultrastructural changes identified via electron microscopy indicate disorganized and destabilized sarcomeres [67,82], which could significantly impair contractile function within the cardiac muscle.
In addition to total loss of sarcomeric proteins and sarcomere ultrastructural disarray, increased fibrosis and a decreased ratio of α/β-MHC may also have a role in impaired cardiac function in cachexia. Fibrosis, associated with a noncompliant heart that impedes relaxation and diastolic function [83], has been found in multiple cachexia studies [64,67,74,84]. Although significant fibrosis likely contributes to impaired diastolic function in cachexia, fibrosis-independent mechanisms are also involved, as isolated cardiac myocytes freed from fibrotic lesions also exhibit slow relaxation properties [65,74]. Fetal gene reactivation in heart failure is associated with a decrease in the “fast” α-MHC isoform and an increase in the “slow” β-MHC isoform. Both mRNA and protein analyses reveal a decreased ratio of α/β-MHC in hearts of C26 tumor-bearing mice [66,67], similar to other causes of heart failure in humans [85,86]. β-MHC exhibits slower cross-bridge kinetics, decreasing maximum shortening velocity and peak contraction compared with α-MHC. Interestingly, the change in contractility resulting from the MHC isoform switch is calcium independent [87,88], suggesting that both calcium-dependent [74] and calcium-independent mechanisms may be contributing to impaired contractile function.
Muscle size, including cardiac muscle, is controlled by the relative rates of protein synthesis and degradation. Upregulation of UPS, ALP, and apoptosis are known to promote cancer cachexia [89,90]. The hyperactivity of these pathways leads to muscle atrophy. Various pre-clinical studies exploring cancer-induced cardiac muscle wasting are at odds regarding which proteolytic pathway is predominant in the heart during cancer cachexia. Some work suggests that upregulation of the UPS and apoptosis/caspase proteolytic systems may be involved in the loss of myofibrillar protein in cachectic hearts [48,65,66,91,92]. Others deem cardiac muscle wasting the result of increased activation of the ALP [71,84,93]. It is possible that alterations to any of these three pathways may lead to cardiac muscle wasting in the cachectic heart, and different mechanisms of proteolysis may be predominant at different stages of cachexia and with different models of cachexia. In addition to increased proteolysis, a decrease in protein synthesis also contributes to the net loss of cardiac muscle protein in cachexia. The protein synthesis rate in the hearts of C57BL/6 mice with ApcMin/+ induced cancer cachexia was found to be reduced by approximately 70% compared to control mice [93]. Similarly, cardiac protein synthesis was reduced in LLC tumor-bearing male mice compared to control mice [69]. Taken together, both increased protein degradation and decreased protein synthesis have been observed in the hearts of cachectic animals. Thus, net protein balance in the cachectic heart disproportionately favors catabolism.

4. Metabolic Remodeling

Metabolic changes, including inefficient energy production, increased metabolic rate, and dysregulation of metabolic pathways, are paramount in the development and progression of cachexia-induced skeletal muscle wasting. Underlying many of these changes is a profound decrease in the function of mitochondria, which are responsible for approximately 90% of the body’s energy production. Due to the high energetic requirements of cardiac muscle, it seems reasonable to suggest that metabolic aberrations in cachexia likely have an important role in the pathogenesis of cachexia-induced cardiomyopathy as well.

4.1. Clinical Observations of Cardiac Metabolic Changes

Clinically, little is known about changes in cardiac metabolism in cancer cachexia, but limited evidence suggests an increased energy expenditure and altered substrate utilization in cardiac tissue. Increased resting heart rate has been noted in cachectic patients, and although not necessarily causal, this was significantly correlated with body weight-adjusted resting energy expenditure [94]. Indeed, cardiac energy expenditure accounts for approximately 10% of resting metabolic rate in healthy adults [95], meaning increased heart rate may well contribute to increased whole body energy utilization. Treatment of weight-losing cancer patients with propranolol (beta-adrenergic receptor antagonist) led to a decrease in resting energy expenditure compared to treatment with indomethacin (prostaglandin synthesis inhibition), morphine (pain reliever), or placebo, implicating increased sympathetic nervous system activity as a contributor to increased energy expenditure in weight-losing cancer [59]. In a crossover study, atenolol (beta-1 antagonist) and propranolol (beta-1/2 antagonist) decreased REE by 6% (77 kcal/day) and 4% (48 kcal/day), respectively, and this was associated with decreased heart rate, oxygen consumption, and CO2 expiration [47]. In addition to increased heart rate, changes in energy substrate availability and energy production efficiency may also contribute to increased energy utilization in cardiac tissue. Different types of cancer are associated with different cardiac glucose uptake rates measured by 18F-FDG positron emission tomography CT scans [60], which changes the ratio of glucose to fatty acids being oxidized, and subsequently the oxygen requirements in cardiac tissue. A summary of these studies identifying metabolic changes is found in Table 1.
Metabolic aberrations leading to increased energy expenditure [18,20,94] and increased futile cycling [25,26,27] are key components of cachexia-induced skeletal muscle wasting and may therefore also have a role in cardiac atrophy and dysfunction. Although little is known clinically in this regard, pre-clinical work is beginning to uncover evidence of metabolic dysfunction in the cachectic heart.

4.2. Mechanistic Contributors to Cardiac Metabolic Changes

The healthy heart is a metabolically demanding organ that relies primarily on mitochondrial metabolism to provide the necessary ATP for contraction, relaxation, and the maintenance of resting membrane potential. It is estimated that the heart consumes approximately six kilograms of ATP per day [96]. To support this large metabolic demand, mitochondria make up between 30–40% of the volume of cardiac myocytes and produce approximately 95% of the ATP [97]. Beyond energy production, mitochondria also play a pivotal role in regulating cellular redox balance and are a primary site of reactive oxygen species production [98]. Therefore, this section on mechanisms of metabolic remodeling will highlight cancer-induced mitochondrial alterations in key mitochondrial-related measurements from animal models. Figure 2 presents an overview of findings related to mitochondrial health and function.

4.2.1. Substrate Utilization

It is well established that the healthy, normal heart preferentially uses fatty acid β-oxidation over carbohydrate sources (i.e., glucose and lactate) for ATP production. Isotope tracer studies estimated that ∼60–90% of the acetyl-CoA comes from β-oxidation of fatty acids [99] and 10–40% comes from the oxidation of pyruvate in the form of glucose or lactate [99]. To date, the data on the effect of cancer on cardiac substrate utilization are equivocal. An early study by Drott et al. demonstrated that hearts from tumor-bearing rats perfused in an ex vivo working heart preparation had decreased glucose uptake compared to fed and fasted control rats, which suggests either decreased capacity for glucose uptake into cells or a lower rate of glucose oxidation [100]. Addition of palmitate to the media of a cell culture model of cachexia caused increased maximal oxygen consumption and upregulation of several genes related to lipid metabolism [63], lending support to the ex vivo heart findings from Drott et al. In contrast, Lee et al. indirectly estimated oxidative vs. glycolytic flux using two-photon excitation fluorescence to assess the redox ratio between FAD and NADH in hearts from cachectic mice [101]. A lower redox ratio was found in this study, indicating decreased utilization of mitochondrial oxidative metabolism and a greater reliance on glycolysis. In support of this finding, proteomic profiling identified downregulation of several mitochondrial electron transport chain proteins [82]. One potential explanation for this finding was the potential for mild hypoxia. HIF1α protein, a transcription factor activated by hypoxia, was significantly elevated in hearts from tumor-bearing animals [101,102]. These studies used HIF1α as a marker of hypoxia, which, in the context of decreased venous oxygen concentration [102], increased skeletal muscle HIF1α [102], and decreased skeletal muscle capillary density [103], suggests impaired oxygen delivery to tissues. Together, these studies implicate hypoxia as a potential contributor to cachexia, which would likely alter substrate utilization.
Unfortunately, substrate utilization in cardiac muscle cannot be inferred by comparison to skeletal muscle because their preferred substrate and regulation are different. For example, cancer-induced limb muscle wasting has been associated with insulin resistance [104,105,106], inhibition of glucose oxidation [107], and excessive fatty acid oxidation [108,109]. Moreover, there is a strong possibility that experimental results are model-dependent. Thackeray et al. [110] compared glucose metabolism in hearts of C26 and B16F10 tumor mice using PET scans for glucose uptake. In the advanced stage, cachectic hearts of C26 mice had elevated glucose uptake, consistent with the findings of Lee et al. [101]. On the other hand, B16F10 mice had lower glucose uptake [110], supporting the discovery of decreased glucose uptake found in ex vivo hearts by Drott et al. [100]. Therefore, further work is necessary to clarify the use of substrates and regulation of oxidative metabolism in the heart, especially in light of suggestions to use pharmacological inhibitors of fatty acid metabolism to treat/prevent skeletal muscle wasting in cachexia [111].

4.2.2. Mitochondrial Morphology

One potential mechanism by which oxidative metabolism may be disrupted by cachexia is through changes in mitochondrial morphology. Electron microscopy images of hearts from multiple cancer cachexia mouse models reveal lower mitochondrial density, randomly dispersed and swollen mitochondria, impaired membrane integrity, and disorganized cristae [67,69,82,84]. Although not systematically studied, we speculate these structural changes to the inner mitochondrial membrane and cristae structure may potentially reduce oxidative phosphorylation capacity [112]. As is well-known, the cristae contain the electron transport chain (ETC) complexes. Currently, it is thought that the distance between these proteins and the cristae dramatically alters the rate of electron transfer through the ETC, and thus the rate of ATP production. For example, in highly oxidative tissue such as the heart, the cristae are closely stacked and take up most of the mitochondrial volume [113]. In less metabolically active tissue, such as the liver, the cristae are further apart [112]. Indeed, isolated mitochondria show condensed cristae when actively respiring, with cristae becoming more spread out in the absence of substrate [114].

4.2.3. Mitochondrial Protein Synthesis and Mitophagy

In addition to changes in cristae structure, the protein content of the ETC complexes may also be reduced [64,69]. Like myocyte structural proteins, mitochondrial protein content is determined by the combination of protein synthesis through translation initiation and protein degradation through mitochondrial-specific autophagy (mitophagy). Several groups have demonstrated reduced cardiac protein synthesis [69,93]. It follows that since most mitochondrial proteins are nuclear encoded, reduced cellular protein synthesis would impact the mitochondrial fraction in a similar manner. Contrary to this thought, Lee et al. demonstrated elevated expression of mitochondrial biogenesis proteins (TFAM and TACO1) and mitochondrial-specific translation initiation factor proteins mtIF2 and mtIF3 in LLC tumor-bearing mice [101]. However, this did not lead to differences in mitochondrial protein synthesis in this study or in a recent study by Brown et al. [115]. Unfortunately, the ETC content does not resolve the different findings in these studies. Lee et al. demonstrated increases in ETC complex II and III, while Berent et al. [69] and Fernandes et al. [64] showed decreases in some of the complexes. These findings suggest that mitochondrial protein synthesis may be variable, and reduced mitochondrial volume [116] may be regulated by protein degradation more than protein synthesis.
The aforementioned structural changes and loss of membrane potential [116] in cachectic hearts would suggest that elevated degradation of mitochondria is required to maintain a healthy mitochondria population. Mitophagy, the selective form of macroautophagy (commonly referred to as autophagy), is capable of removing mitochondria. Unlike proteins in the cell that can be degraded by calpain or the UPS [117], the large organelle structure of mitochondria requires a process that can degrade lipids, protein, and DNA. This process is accomplished through the targeting of mitochondria and directed formation of a double membrane autophagosome, fusion with a lysosome, and degradation by the cathepsin family of proteases (reviewed in [118,119]). The majority of the autophagosome is consumed in the process of mitochondrial degradation and is not recycled, which makes accurate measurement of autophagy in frozen tissue samples challenging. Despite this, there are several markers which have been consistently associated with increased autophagy and commonly measured in the field. One such marker of autophagy is the ratio of microtubule-associated protein light chain 3 (LC3) II to LC3I (LC3II/LC3I) [120]. Several studies demonstrate an increase in the LC3 ratio in hearts of tumor-bearing animals [68,72,84,110,121]. These findings suggest that autophagy is increased in cardiac myocytes during cachexia and may lead to increased rates of protein degradation of mitochondria.
One of the more robust findings in cachexia literature is the increase in expression of BNIP3, a protein that is associated with the mitochondrial membrane and plays an important role in targeting degenerated mitochondria for mitophagy. A total of six papers found elevated BNIP3 mRNA or protein expression in cachectic hearts [62,64,68,72,121,122]. Only one study did not show an increase in BNIP3, and interestingly this study did not show cardiac atrophy or contractile dysfunction [123]. These findings are supported by electron microscopy images showing a higher number of double-membraned autophagic vacuoles in late-stage atrophic hearts [84]. A question that these data pose is whether mitophagy is beneficial or damaging to the heart? Is it specifically targeting damaged mitochondria in an attempt to make the overall population of mitochondria in the atrophied heart healthier? Or is the degradation non-specific and contributing to disorganized mitochondrial morphology and leading to mitochondrial dysfunction?

4.2.4. Mitochondrial Respiration

Mitochondrial function is commonly defined by either respiration or reactive oxygen species production (ROS). First, mitochondrial respiration is estimated by oxygen consumption using modified definitions from Chance and Williams [124]. Briefly, carbon providing substrates (i.e., malate, pyruvate, succinate, glutamate, or palmitoylcarnitine) are added to isolated mitochondria or permeabilized fibers. Then, ADP is added as a substrate to stimulate a near maximal rate of respiration, classically defined as state 3 respiration. The second common measurement is state 4 respiration, which is either measured following ADP depletion or by the addition of oligomycin, an inhibitor of ATP synthase activity. One common interpretation of these data is to look at the respiratory control ratio (RCR) that is calculated by dividing state 3 by state 4. In theory, a healthy mitochondrion would have a high rate of respiration when stimulated by ADP and a low rate of respiration when ATP synthase is inhibited, which would create a high ratio of state 3 to state 4. Note that there are a variety of protocols for specific evaluation of substrates and electron transport chain activity covered in depth by Gnagier [125].
Smuder et al. reported impaired mitochondrial function in permeabilized cardiac myocytes from animals with C26 tumors [68]. These data demonstrated that cachectic hearts had lower state 3 respiration, higher state 4 respiration, and therefore a lower RCR compared to control animals. To our knowledge, this is the only study that has measured these parameters in an isolated ex vivo model. The other studies that have evaluated mitochondrial respiration have done so by using the previously mentioned method with cardiac myocytes cultured in conditioned medium from tumor cell culture. The conditioned medium was used to model the inflammatory cytokines released from the tumor that likely significantly contribute to cardiac atrophy. Berent et al. [69] did not find a difference in respiration in these cells in either state 3 or state 4 (listed as basal and leak, respectively), but did find a lower maximal rate of respiration, likely caused by the measured differences in ETC complexes. In a similar model, Nukaga et al. [116] added ascites from tumor animals in the cell culture media that lead to a reduced change in state 3 (basal), but no change in state 4 (leak). Although not calculated, these numbers would lead to a decreased RCR. Lee et al. also used this model and showed similar findings to Berent et al. [69], where there were no changes in basal or leak, but a lower maximal respiration capacity [101]. In contrast, Shäfer et al. demonstrated higher basal and leak respiration in cultured myocytes [63]. It is important to note that all these cell culture measurements were collected using the mitochondrial stress kit by Seahorse Bioscience, and there is still a debate on the interpretation and/or physiological relevance of these markers [126]. Additionally, all the cell culture studies were conducted with either immortalized cell lines (AC16 and H9c2) or primary neonatal rat cardiac myocytes, which have different characteristics compared to primary adult cardiac myocytes. Furthermore, cells were not paced in culture which drastically changes metabolic demand compared to the in vivo environment. Further studies using paced, adult cardiac myocytes may clarify discrepancies between studies.

4.2.5. Oxidative Stress

In addition to alterations in mitochondrial respiration, the mitochondria can also contribute to the production of ROS, leading to oxidative stress that has been strongly associated with cachexia [127]. In skeletal muscle, mitochondria ROS production is increased before overt signs of cachexia such as muscle atrophy or changes in body weight occur [24,128]. Oxidative stress affects a multitude of pathways that may, in part, lead to the cardiac atrophy observed in cancer cachexia. Indeed, mitochondrial ROS can increase protein degradation, reduce protein synthesis, and reduce mitochondrial respiration [117].
Direct, in vivo measurement of ROS is technically difficult and therefore is commonly estimated through the measurement of markers of oxidative stress or in ex vivo approaches that use fluorogenic probes [129]. Markers of oxidative stress are reviewed in detail by Frijhoff et al. [130], but in general these markers measure stable by-products formed under redox stress. Examples include protein carbonyls formed through the oxidative cleavage of protein backbones, lipid oxidation products generated by H-atom abstraction, and DNA oxidation. Relatively limited evidence supports the notion that cancer leads to elevated oxidative damage to DNA through the measurement of deoxyguanosine (8-OHdG) [131,132]. Furthermore, TBARS, a marker of lipid peroxidation, was increased with no difference in protein carbonyl formation [133].
Two studies have used fluorogenic probes to provide high sensitivity and specificity real-time measurements for ROS. Mitochondrial hydrogen peroxide production was higher in hearts from tumor-bearing animals compared to control animals [68]. The use of a mitochondrial targeted antioxidant (SS-31) was able to prevent oxidative stress and restore function. Similarly, a mitochondrial-specific fluorogenic dye (MitoSOX) in cardiac myocytes cultured with media from cancer cells demonstrated a higher mitochondrial superoxide production [101]. These data are supported by unbiased RNA sequencing and pathway analysis data that revealed many differentially expressed genes related to oxidative stress [134]. In general, the volume of data is not strong enough to definitively state oxidative stress is present in cachectic hearts. However, the few studies that exist support the notion that cardiac muscle experiences oxidative stress in response to tumors, and the mitochondria are likely a significant source of ROS.
Whether or not oxidative stress is a cause of atrophy has long been debated in skeletal muscle atrophy models [135]. This mechanism has not been systematically studied in heart muscle. However, the best evidence is the indirect data using exogenous antioxidants to reduce oxidative stress. To date, four studies have been completed using supplements that have antioxidant properties in cells. Resveratrol, a polyphenol with strong ROS scavenging properties [136], prevented cardiac atrophy and dysfunction in the C26 mouse model [137]. Oxypurinol, which acts as an antioxidant by inhibiting xanthine oxidase ROS production, provided some protection against cardiac atrophy and contractile dysfunction in the Yoshida AH-130 hepatoma cancer cachexia model [138]. Neither of these studies measured markers of oxidative stress, sites of ROS production, or antioxidant capacity, which limits the ability to attribute these beneficial effects to the antioxidant properties. In cachexia cell culture, the addition of MitoTemo, a mitochondrial-specific antioxidant, was able to prevent increased superoxide concentrations and improve mitochondrial function and cell viability to a hypoxia challenge assay [101]. The mitochondrial-specific antioxidant SS-31 prevented increases in mitochondrial ROS production, and improved mitochondrial function and cardiac function, but did not demonstrate robust protection against cardiac atrophy [68].

4.2.6. Future Research

There are several questions that remain to be answered about the mechanisms of altered bioenergetics and metabolism in cardiac cachexia. As the data show, substrate selection by the heart needs to be further explored and considered across a variety of models, especially in light of the opposite findings between models in the Thackeray study [110]. Translating these findings may lead to nutritional and pharmacological interventions to improve cardiac energy production and metabolism in cancer cachexia.
Mitochondrial physiology appears to play a prominent role in cardiac cachexia and may be a potential therapeutic target. To date, the field lacks well-controlled studies using genetic models to isolate specific pathways. Treatments with antioxidants and inhibitors have demonstrated potential, however, care should be taken to understand if these molecules increase tumor burden. Of note, none of the studies presented here [68,137,138] reported increased tumor growth. However, many exogenous supplements [139,140,141] and even tissue-specific genetic manipulation [142] can increase tumor growth rate, making tumor growth an important consideration moving forward.

5. Conclusions

The development and progression of cancer-induced cachexia involves a complex interplay between tumor- and host-derived factors that involves multiple tissues and organs [34]. One of the organs involved in this syndrome is the heart. Although atrophy of the cardiac muscle in cachectic patients was observed decades ago [78], we are just beginning to understand the underlying mechanisms of cachexia-induced cardiomyopathy. Increasing evidence of cardiac functional, structural, and metabolic changes in patients with cancer cachexia suggests the heart is significantly affected by, and also contributes to, cachexia pathophysiology, making the heart a vital area of cachexia research. Figure 3 presents an overall summary of the findings.
Cachexia-induced cardiomyopathy has both similarities and differences compared with other types of heart failure, which affects the clinical management of this condition. Both clinical and pre-clinical studies have identified impaired systolic function [54,67], with some work also identifying impaired diastolic function [48,74]. Decreased calcium cycling [74], increased fibrotic development [48,67], and changes in myosin isoform expression [66,67] may all contribute to functional deficits and are all common to multiple cardiomyopathy etiologies. However, the atrophic structural changes are in stark contrast to either the hypertrophic or dilated phenotypes characteristic of many other cardiomyopathies [35,143,144]. Decreased total expression of sarcomeric proteins [66], decreased cardiac myocyte width in the absence of increased length [74], and thinning of the ventricular walls [48,66] suggests an imbalance between proteolysis and protein synthesis, similar to cachectic skeletal muscle. Atrophy may be a contributor to the overall decrease in force production in the cachectic heart [52], although additional structural and morphological changes such as t-tubule remodeling occurring in conjunction with decreased sarcomeric protein expression may also contribute and should be investigated [145]. Furthermore, similar to other forms of heart failure, significant alterations in whole body and cardiac metabolism may hinder energy-requiring functions in the heart, such as maintenance of electrochemical gradients and intracellular calcium concentrations, and cross-bridge cycling. Considerable evidence exists for cachexia-induced skeletal muscle mitochondrial dysfunction, with increasing evidence showing mitochondrial abnormalities in the heart as well [68,101]. As the heart has a high metabolic demand, contributing significantly to basal metabolic rate [95], decreased energy production by cardiac mitochondria may contribute to both functional deficits and atrophy of the heart muscle.
Published literature related to the cardiac effects of cancer cachexia reviewed in this paper identifies several consistent findings. First, in vivo systolic dysfunction (decreased contractility) was found in most pre-clinical studies undertaking echocardiographic measurements [48,65,67], and this was corroborated by limited clinical data also examining cardiac function in cachectic patients [46,54,56]. Second, cardiac atrophy was a consistent finding in many pre-clinical models [48,67,71,81] as well as some studies in humans [48,57]. Finally, evidence of autophagy was significantly and consistently increased in numerous pre-clinical models [68,72,121]. These findings provide important inroads into our understanding of cachexia-induced cardiomyopathy. However, many questions remain related to the mechanistic underpinnings of cachexia-induced cardiomyopathy. Specifically, there are several areas in which existing studies present contradictory findings. Related to cardiac function, some evidence suggests that isolated heart and cardiac myocyte functional deficits underlie in vivo dysfunction [74]; however, not all studies are in agreement [76]. Related to metabolism and mitochondrial function, further work is needed to understand changes in substrate utilization, mitochondrial respiration, and mitochondrial protein synthesis. In addition to understanding contradictory findings in existing literature, there are other areas of research that should be considered. First, identification of cardiac changes early in cachexia pathogenesis will enable identification of biomarkers to predict cardiomyopathy risk, as well as enable targeted therapeutic development [24]. Second, understanding the reversibility of cachexia-induced cardiac dysfunction in patients who are living with stable disease or are in remission will be vital to development of long-term management strategies.
Research progress surrounding cachexia-induced cardiomyopathy is not without challenges. Clinically, identification of cachexia-specific effects on the heart is difficult to ascertain. The type and stage of the tumor, cancer therapies, and pre-existing cardiovascular disease are all factors that complicate the interpretation of clinical data. Although animal models have similarities with the human condition, the high tumor burden, young age, non-metastatic disease, and rapidity of cachexia development in the most commonly used models pose challenges to clinical translatability [146,147]. Finally, mice have differences in cardiac structure and function compared to humans [148,149], necessitating care in the interpretation and extrapolation of pre-clinical findings to humans. Nevertheless, the parallel nature of clinical and pre-clinical research in cachexia-induced cardiomyopathy has provided important inroads into understanding this complex condition.
In conclusion, cachexia-induced cardiomyopathy is characterized by impaired in vivo systolic function, significant atrophy of the cardiac muscle, and evidence of energy production and mitochondrial functional deficits. Because cardiac insufficiency can lead to muscle wasting independent of cancer [35], cardiac dysfunction in cachexia may contribute to cachexia pathophysiology. Indeed, many symptoms, including fatigue, exercise intolerance, and dyspnea, are shared between cancer cachexia and heart failure [36]. Therefore, understanding the effects of cancer cachexia on the heart is imperative for optimal clinical management and improved treatment options for patients with cachexia-induced cardiomyopathy.

Author Contributions

Conceptualization, M.L.L.; Writing—Original Draft Preparation, M.P.W., A.G.B., M.L.L.; Writing—Review and Editing, M.P.W., A.G.B., M.L.L. All authors have read and agreed to the published version of the manuscript.

Funding

This paper received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Anker, M.S.; Holcomb, R.; Muscaritoli, M.; von Haehling, S.; Haverkamp, W.; Jatoi, A.; Morley, J.E.; Strasser, F.; Landmesser, U.; Coats, A.J.S.; et al. Orphan Disease Status of Cancer Cachexia in the USA and in the European Union: A Systematic Review. J. Cachexia Sarcopenia Muscle 2019, 10, 22–34. [Google Scholar] [CrossRef]
  2. Tisdale, M.J. Cachexia in Cancer Patients. Nat. Rev. Cancer 2002, 2, 862–871. [Google Scholar] [CrossRef]
  3. Baracos, V.E.; Martin, L.; Korc, M.; Guttridge, D.C.; Fearon, K.C.H. Cancer-Associated Cachexia. Nat. Rev. Dis. Primers 2018, 4, 17105. [Google Scholar] [CrossRef]
  4. Sun, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  5. Ferlay, J.; Ervik, M.; Lam, F.; Colombet, M.; Mery, L.; Pineros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today; International Agency for Research on Cancer: Lyon, France. Available online: https://gco.iarc.fr/today (accessed on 20 October 2021).
  6. Fearon, K.; Strasser, F.; Anker, S.D.; Bosaeus, I.; Bruera, E.; Fainsinger, R.L.; Jatoi, A.; Loprinzi, C.; MacDonald, N.; Mantovani, G.; et al. Definition and Classification of Cancer Cachexia: An International Consensus. Lancet Oncol. 2011, 12, 489–495. [Google Scholar] [CrossRef]
  7. Prado, C.M.M.; Lieffers, J.R.; McCargar, L.J.; Reiman, T.; Sawyer, M.B.; Martin, L.; Baracos, V.E. Prevalence and Clinical Implications of Sarcopenic Obesity in Patients with Solid Tumours of the Respiratory and Gastrointestinal Tracts: A Population-Based Study. Lancet Oncol. 2008, 9, 629–635. [Google Scholar] [CrossRef]
  8. Martin, L.; Birdsell, L.; MacDonald, N.; Reiman, T.; Clandinin, M.T.; McCargar, L.J.; Murphy, R.; Ghosh, S.; Sawyer, M.B.; Baracos, V.E. Cancer Cachexia in the Age of Obesity: Skeletal Muscle Depletion Is a Powerful Prognostic Factor, Independent of Body Mass Index. J. Clin. Oncol. 2013, 31, 1539–1547. [Google Scholar] [CrossRef]
  9. Li, Z.; Zhu, L.; Zheng, H.; Jiang, W.; Wang, Y.; Jiang, Z.; Xu, J. Serum IL-35 Levels Is a New Candidate Biomarker of Cancer-related Cachexia in Stage IV Non-small Cell Lung Cancer. Thorac. Cancer 2022, 13, 716–723. [Google Scholar] [CrossRef]
  10. Ross, P.J.; Ashley, S.; Norton, A.; Priest, K.; Waters, J.S.; Eisen, T.; Smith, I.E.; O’Brien, M.E.R. Do Patients with Weight Loss Have a Worse Outcome When Undergoing Chemotherapy for Lung Cancers? Br. J. Cancer 2004, 90, 1905–1911. [Google Scholar] [CrossRef]
  11. Wallengren, O.; Lundholm, K.; Bosaeus, I. Diagnostic Criteria of Cancer Cachexia: Relation to Quality of Life, Exercise Capacity and Survival in Unselected Palliative Care Patients. Supportive Care Cancer 2013, 21, 1569–1577. [Google Scholar] [CrossRef]
  12. Gourin, C.G.; Couch, M.E.; Johnson, J.T. Effect of Weight Loss on Short-Term Outcomes and Costs of Care after Head and Neck Cancer Surgery. Ann. Otol. Rhinol. Laryngol. 2014, 123, 101–110. [Google Scholar] [CrossRef] [PubMed]
  13. Arthur, S.T.; van Doren, B.A.; Roy, D.; Noone, J.M.; Zacherle, E.; Blanchette, C.M. Cachexia among US Cancer Patients. J. Med. Econ. 2016, 19, 874–880. [Google Scholar] [CrossRef] [PubMed]
  14. Wakabayashi, H.; Arai, H.; Inui, A. The Regulatory Approval of Anamorelin for Treatment of Cachexia in Patients with Non-Small Cell Lunger Cancer, Gastric Cancer, and Colorectal Cancer in Japan: Facts and Numbers. J. Cachexia Sarcopenia Muscle 2021, 12, 14–16. [Google Scholar] [CrossRef]
  15. Roeland, E.J.; Bohlke, K.; Baracos, V.E.; Bruera, E.; del Fabbro, E.; Dixon, S.; Fallon, M.; Herrstedt, J.; Lau, H.; Platek, M.; et al. Management of Cancer Cachexia: ASCO Guideline. J. Clin. Oncol. 2020, 38, 2438–2453. [Google Scholar] [CrossRef]
  16. Dhanapal, R.; Saraswathi, T.; Rajkumar, G. Cancer Cachexia. J. Oral Maxillofac. Pathol. 2011, 15, 257. [Google Scholar] [CrossRef]
  17. Prado, C.M.; Laviano, A.; Gillis, C.; Sung, A.D.; Gardner, M.; Yalcin, S.; Dixon, S.; Newman, S.M.; Bastasch, M.D.; Sauer, A.C.; et al. Examining Guidelines and New Evidence in Oncology Nutrition: A Position Paper on Gaps and Opportunities in Multimodal Approaches to Improve Patient Care. Supportive Care Cancer 2022, 30, 3073–3083. [Google Scholar] [CrossRef]
  18. Lieffers, J.R.; Mourtzakis, M.; Hall, K.D.; McCargar, L.J.; Prado, C.M.M.; Baracos, V.E. A Viscerally Driven Cachexia Syndrome in Patients with Advanced Colorectal Cancer: Contributions of Organ and Tumor Mass to Whole-Body Energy Demands. Am. J. Clin. Nutr. 2009, 89, 1173–1179. [Google Scholar] [CrossRef] [Green Version]
  19. Zhang, G.; Liu, Z.; Ding, H.; Zhou, Y.; Doan, H.A.; Sin, K.W.T.; Zhu, Z.J.; Flores, R.; Wen, Y.; Gong, X.; et al. Tumor Induces Muscle Wasting in Mice through Releasing Extracellular Hsp70 and Hsp90. Nat. Commun. 2017, 8, 589. [Google Scholar] [CrossRef] [Green Version]
  20. Friesen, D.E.; Baracos, V.E.; Tuszynski, J.A. Modeling the Energetic Cost of Cancer as a Result of Altered Energy Metabolism: Implications for Cachexia. Theor. Biol. Med. Model. 2015, 12, 17. [Google Scholar] [CrossRef] [Green Version]
  21. Lerner, L.; Tao, J.; Liu, Q.; Nicoletti, R.; Feng, B.; Krieger, B.; Mazsa, E.; Siddiquee, Z.; Wang, R.; Huang, L.; et al. MAP3K11/GDF15 Axis Is a Critical Driver of Cancer Cachexia. J. Cachexia Sarcopenia Muscle 2016, 7, 467–482. [Google Scholar] [CrossRef]
  22. Loumaye, A.; de Barsy, M.; Nachit, M.; Lause, P.; Frateur, L.; van Maanen, A.; Trefois, P.; Gruson, D.; Thissen, J.P. Role of Activin A and Myostatin in Human Cancer Cachexia. J. Clin. Endocrinol. Metab. 2015, 100, 2030–2038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Deans, C.; Wigmore, S.J. Systemic Inflammation, Cachexia and Prognosis in Patients with Cancer. Curr. Opin. Clin. Nutr. Metab. Care 2005, 8, 265–269. [Google Scholar] [CrossRef] [PubMed]
  24. Brown, J.L.; Rosa-Caldwell, M.E.; Lee, D.E.; Blackwell, T.A.; Brown, L.A.; Perry, R.A.; Haynie, W.S.; Hardee, J.P.; Carson, J.A.; Wiggs, M.P.; et al. Mitochondrial Degeneration Precedes the Development of Muscle Atrophy in Progression of Cancer Cachexia in Tumour-Bearing Mice. J. Cachexia Sarcopenia Muscle 2017, 8, 926–938. [Google Scholar] [CrossRef] [PubMed]
  25. Han, J.; Meng, Q.; Shen, L.; Wu, G. Interleukin-6 Induces Fat Loss in Cancer Cachexia by Promoting White Adipose Tissue Lipolysis and Browning. Lipids Health Dis. 2018, 17, 14. [Google Scholar] [CrossRef] [Green Version]
  26. Kir, S.; White, J.P.; Kleiner, S.; Kazak, L.; Cohen, P.; Baracos, V.E.; Spiegelman, B.M. Tumour-Derived PTH-Related Protein Triggers Adipose Tissue Browning and Cancer Cachexia. Nature 2014, 513, 100–104. [Google Scholar] [CrossRef]
  27. Kliewer, K.L.; Ke, J.Y.; Tian, M.; Cole, R.M.; Andridge, R.R.; Belury, M.A. Adipose Tissue Lipolysis and Energy Metabolism in Early Cancer Cachexia in Mice. Cancer Biol. Ther. 2015, 16, 886–897. [Google Scholar] [CrossRef] [Green Version]
  28. Cao, D.; Wu, G.; Zhang, B.; Quan, Y.; Wei, J.; Jin, H.; Jiang, Y.; Yang, Z. Resting Energy Expenditure and Body Composition in Patients with Newly Detected Cancer. Clin. Nutr. 2010, 29, 72–77. [Google Scholar] [CrossRef]
  29. Falconer, J.S.; Fearon, K.C.H.; Plester, C.E.; Ross, J.A.; Carter, D.C. Cytokines, the Acute-Phase Response, and Resting Energy Expenditure in Cachectic Patients with Pancreatic Cancer. Ann. Surg. 1994, 219, 325–331. [Google Scholar] [CrossRef]
  30. Argilés, J.M.; Busquets, S.; Stemmler, B.; López-Soriano, F.J. Cancer Cachexia: Understanding the Molecular Basis. Nat. Rev. Cancer 2014, 14, 754–762. [Google Scholar] [CrossRef]
  31. Smith, K.L.; Tisdale, M.J. Increased Protein Degradation and Decreased Protein Synthesis in Skeletal Muscle during Cancer Cachexia. Br. J. Cancer 1993, 67, 680–685. [Google Scholar] [CrossRef] [Green Version]
  32. Sandri, M. Protein Breakdown in Cancer Cachexia. Semin. Cell Dev. Biol. 2016, 54, 11–19. [Google Scholar] [CrossRef] [PubMed]
  33. Bossola, M.; Marzetti, E.; Rosa, F.; Pacelli, F. Skeletal Muscle Regeneration in Cancer Cachexia. Clin. Exp. Pharmacol. Physiol. 2016, 43, 522–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Argilés, J.M.; Stemmler, B.; López-Soriano, F.J.; Busquets, S. Inter-Tissue Communication in Cancer Cachexia. Nat. Rev. Endocrinol. 2018, 15, 9–20. [Google Scholar] [CrossRef]
  35. von Haehling, S.; Ebner, N.; dos Santos, M.R.; Springer, J.; Anker, S.D. Muscle Wasting and Cachexia in Heart Failure: Mechanisms and Therapies. Nat. Rev. Cardiol. 2017, 14, 323–341. [Google Scholar] [CrossRef] [PubMed]
  36. Kazemi-Bajestani, S.M.R.; Becher, H.; Fassbender, K.; Chu, Q.; Baracos, V.E. Concurrent Evolution of Cancer Cachexia and Heart Failure: Bilateral Effects Exist. J. Cachexia Sarcopenia Muscle 2014, 5, 95–104. [Google Scholar] [CrossRef]
  37. Alexandre, J.; Cautela, J.; Ederhy, S.; Damaj, G.L.; Salem, J.; Barlesi, F.; Farnault, L.; Charbonnier, A.; Mirabel, M.; Champiat, S.; et al. Cardiovascular Toxicity Related to Cancer Treatment: A Pragmatic Approach to the American and European Cardio-Oncology Guidelines. J. Am. Heart Assoc. 2020, 9, e018403. [Google Scholar] [CrossRef] [PubMed]
  38. Beavers, C.J.; Rodgers, J.E.; Bagnola, A.J.; Beckie, T.M.; Campia, U.; di Palo, K.E.; Okwuosa, T.M.; Przespolewski, E.R.; Dent, S. Cardio-Oncology Drug Interactions: A Scientific Statement from the American Heart Association. Circulation 2022, 145, e811–e838. [Google Scholar] [CrossRef]
  39. Kostakou, P.M.; Kouris, N.T.; Kostopoulos, V.S.; Damaskos, D.S.; Olympios, C.D. Cardio-Oncology: A New and Developing Sector of Research and Therapy in the Field of Cardiology. Heart Fail. Rev. 2019, 24, 91–100. [Google Scholar] [CrossRef]
  40. 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]
  41. Ambrus, J.L.; Ambrus, C.M.; Mink, I.B.; Pickren, J.W. Causes of Death in Cancer Patients. J. Med. 1975, 6, 61–64. [Google Scholar]
  42. Inagaki, J.; Rodriguez, V.; Bodey, G.P. Causes of Death in Cancer Patients. Cancer 1974, 33, 568–573. [Google Scholar] [CrossRef]
  43. Kikuchi, K.; Poss, K.D. Cardiac Regenerative Capacity and Mechanisms. Annu. Rev. Cell Dev. Biol. 2012, 28, 719–741. [Google Scholar] [CrossRef] [Green Version]
  44. Lee, D.H.; Park, S.; Lim, S.M.; Lee, M.K.; Giovannucci, E.L.; Kim, J.H.; Kim, S.I.; Jeon, J.Y. Resting Heart Rate as a Prognostic Factor for Mortality in Patients with Breast Cancer. Breast Cancer Res. Treat. 2016, 159, 375–384. [Google Scholar] [CrossRef]
  45. Anker, M.S.; Ebner, N.; Hildebrandt, B.; Springer, J.; Sinn, M.; Riess, H.; Anker, S.D.; Landmesser, U.; Haverkamp, W.; von Haehling, S. Resting Heart Rate Is an Independent Predictor of Death in Patients with Colorectal, Pancreatic, and Non-Small Cell Lung Cancer: Results of a Prospective Cardiovascular Long-Term Study. Eur. J. Heart Fail. 2016, 18, 1524–1534. [Google Scholar] [CrossRef]
  46. Cramer, L.; Hildebrandt, B.; Kung, T.; Wichmann, K.; Springer, J.; Doehner, W.; Sandek, A.; Valentova, M.; Stojakovic, T.; Scharnagl, H.; et al. Cardiovascular Function and Predictors of Exercise Capacity in Patients with Colorectal Cancer. J. Am. Coll Cardiol 2014, 64, 1310–1319. [Google Scholar] [CrossRef] [Green Version]
  47. Hyltander, A.; Daneryd, P.; Sandström, R.; Körner, U.; Lundholm, K. β-Adrenoceptor Activity and Resting Energy Metabolism in Weight Losing Cancer Patients. Eur. J. Cancer 2000, 36, 330–334. [Google Scholar] [CrossRef]
  48. Springer, J.; Tschirner, A.; Haghikia, A.; von Haehling, S.; Lal, H.; Grzesiak, A.; Kaschina, E.; Palus, S.; Pötsch, M.; von Websky, K.; et al. Prevention of Liver Cancer Cachexia-Induced Cardiac Wasting and Heart Failure. Eur. Heart J. 2014, 35, 932–941. [Google Scholar] [CrossRef]
  49. Cunha, G.J.L.; Rocha, B.M.L.; Menezes Falcão, L. Iron Deficiency in Chronic and Acute Heart Failure: A Contemporary Review on Intertwined Conditions. Eur. J. Intern. Med. 2018, 52, 1–7. [Google Scholar] [CrossRef]
  50. Coats, A.J.S.; Clark, A.L.; Piepoli, M.; Volterrani, M.; Poole-Wilson, P.A. Symptoms and Quality of Life in Heart Failure: The Muscle Hypothesis. Br. Heart J. 1994, 72 (Suppl. S2), S36–S39. [Google Scholar] [CrossRef] [Green Version]
  51. Giannoni, A.; Aimo, A.; Mancuso, M.; Piepoli, M.F.; Orsucci, D.; Aquaro, G.D.; Barison, A.; De Marchi, D.; Taddei, C.; Cameli, M.; et al. Autonomic, Functional, Skeletal Muscle, and Cardiac Abnormalities are Associated with Increased Ergoreflex Sensitivity in Mitochondrial Disease. Eur. J. Heart Fail. 2017, 19, 1701–1709. [Google Scholar] [CrossRef]
  52. Anker, M.S.; Sanz, A.P.; Zamorano, J.L.; Mehra, M.R.; Butler, J.; Riess, H.; Coats, A.J.S.; Anker, S.D. Advanced Cancer Is Also a Heart Failure Syndrome: A Hypothesis. Eur. J. Heart Fail. 2021, 23, 140–144. [Google Scholar] [CrossRef]
  53. Anker, M.S.; Haehling, S.; Coats, A.J.S.; Riess, H.; Eucker, J.; Porthun, J.; Butler, J.; Karakas, M.; Haverkamp, W.; Landmesser, U.; et al. Ventricular Tachycardia, Premature Ventricular Contractions, and Mortality in Unselected Patients with Lung, Colon, or Pancreatic Cancer: A Prospective Study. Eur. J. Heart Fail. 2021, 23, 145–153. [Google Scholar] [CrossRef] [PubMed]
  54. Kazemi-Bajestani, S.M.R.; Becher, H.; Butts, C.; Basappa, N.S.; Smylie, M.; Joy, A.A.; Sangha, R.; Gallivan, A.; Kavsak, P.; Chu, Q.; et al. Rapid Atrophy of Cardiac Left Ventricular Mass in Patients with Non-Small Cell Carcinoma of the Lung. J. Cachexia Sarcopenia Muscle 2019, 10, 1070–1082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Rosa, G.M.; Gigli, L.; Tagliasacchi, M.I.; di Iorio, C.; Carbone, F.; Nencioni, A.; Montecucco, F.; Brunelli, C. Update on Cardiotoxicity of Anti-Cancer Treatments. Eur. J. Clin. Investig. 2016, 46, 264–284. [Google Scholar] [CrossRef] [Green Version]
  56. Kazemi-Bajestani, S.M.R.; Becher, H.; Butts, C.; Basappa, N.S.; Smylie, M.; Joy, A.A.; Sangha, R.; Gallivan, A.; Chu, Q.; Baracos, V.E. Undiagnosed Cardiac Deficits in Non-Small Cell Carcinoma Patients in the Candidate Population for Anti-Cachexia Clinical Trials. Supportive Care Cancer 2019, 27, 1551–1561. [Google Scholar] [CrossRef]
  57. Barkhudaryan, A.; Scherbakov, N.; Springer, J.; Doehner, W. Cardiac Muscle Wasting in Individuals with Cancer Cachexia. ESC Heart Fail. 2017, 4, 458–467. [Google Scholar] [CrossRef]
  58. Cai, Q.; Mao, Y.; Yang, Q.; Wen, H.; Lv, Y.; Zhang, R. Are Left Ventricular Muscle Area and Radiation Attenuation Associated with Overall Survival in Advanced Pancreatic Cancer Patients Treated with Chemotherapy? Clin. Radiol. 2020, 75, 238-E1–238-E9. [Google Scholar] [CrossRef]
  59. Hyltander, A.; Körner, U.; Lundholm, K.G. Evaluation of Mechanisms behind Elevated Energy Expenditure in Cancer Patients with Solid Tumours. Eur. J. Clin. Investig. 1993, 23, 46–52. [Google Scholar] [CrossRef]
  60. Heckmann, M.B.; Totakhel, B.; Finke, D.; Anker, M.S.; Müller-Tidow, C.; Haberkorn, U.; Katus, H.A.; Lehmann, L.H. Evidence for a Cardiac Metabolic Switch in Patients with Hodgkin’s Lymphoma. ESC Heart Fail. 2019, 6, 824–829. [Google Scholar] [CrossRef] [Green Version]
  61. Devine, R.D.; Eichenseer, C.M.; Wold, L.E. Minocycline Attenuates Cardiac Dysfunction in Tumor-Burdened Mice. J. Mol. Cell. Cardiol. 2016, 100, 35–42. [Google Scholar] [CrossRef] [Green Version]
  62. Stevens, S.C.W.; Velten, M.; Youtz, D.J.; Clark, Y.; Jing, R.; Reiser, P.J.; Bicer, S.; Devine, R.D.; McCarthy, D.O.; Wold, L.E. Losartan Treatment Attenuates Tumor-Induced Myocardial Dysfunction. J. Mol. Cell. Cardiol. 2015, 85, 37–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Schäfer, M.; Oeing, C.U.; Rohm, M.; Baysal-Temel, E.; Lehmann, L.H.; Bauer, R.; Volz, H.C.; Boutros, M.; Sohn, D.; Sticht, C.; et al. Ataxin-10 Is Part of a Cachexokine Cocktail Triggering Cardiac Metabolic Dysfunction in Cancer Cachexia. Mol. Metab. 2016, 5, 67–78. [Google Scholar] [CrossRef] [PubMed]
  64. Fernandes, L.G.; Tobias, G.C.; Paixão, A.O.; Dourado, P.M.; Voltarelli, V.A.; Brum, P.C. Exercise Training Delays Cardiac Remodeling in a Mouse Model of Cancer Cachexia. Life Sci. 2020, 260, 118392. [Google Scholar] [CrossRef] [PubMed]
  65. Xu, H.; Crawford, D.; Hutchinson, K.R.; Youtz, D.J.; Lucchesi, P.A.; Velten, M.; McCarthy, D.O.; Wold, L.E. Myocardial Dysfunction in an Animal Model of Cancer Cachexia. Life Sci. 2011, 88, 406–410. [Google Scholar] [CrossRef] [Green Version]
  66. Tian, M.; Asp, M.L.; Nishijima, Y.; Belury, M.A. Evidence for Cardiac Atrophic Remodeling in Cancer-Induced Cachexia in Mice. Int. J. Oncol. 2011, 39, 1321–1326. [Google Scholar] [CrossRef]
  67. Tian, M.; Nishijima, Y.; Asp, M.L.; Stout, M.B.; Reiser, P.J.; Belury, M.A. Cardiac Alterations in Cancer-Induced Cachexia in Mice. Int. J. Oncol. 2010, 37, 347–353. [Google Scholar] [CrossRef]
  68. Smuder, A.J.; Roberts, B.M.; Wiggs, M.P.; Kwon, O.S.; Yoo, J.K.; Christou, D.D.; Fuller, D.D.; Szeto, H.H.; Judge, A.R. Pharmacological Targeting of Mitochondrial Function and Reactive Oxygen Species Production Prevents Colon 26 Cancer-Induced Cardiorespiratory Muscle Weakness. Oncotarget 2020, 11, 3502–3514. [Google Scholar] [CrossRef]
  69. Berent, T.E.; Dorschner, J.M.; Meyer, T.; Craig, T.A.; Wang, X.; Kunz, H.; Jatoi, A.; Lanza, I.R.; Chen, H.; Kumar, R. Impaired Cardiac Performance, Protein Synthesis, and Mitochondrial Function in Tumor-Bearing Mice. PLoS ONE 2019, 14, e0226440. [Google Scholar] [CrossRef] [Green Version]
  70. Toledo, M.; Springer, J.; Busquets, S.; Tschirner, A.; López-Soriano, F.J.; Anker, S.D.; Argilés, J.M. Formoterol in the Treatment of Experimental Cancer Cachexia: Effects on Heart Function. J. Cachexia Sarcopenia Muscle 2014, 5, 315–320. [Google Scholar] [CrossRef] [Green Version]
  71. Musolino, V.; Palus, S.; Tschirner, A.; Drescher, C.; Gliozzi, M.; Carresi, C.; Vitale, C.; Muscoli, C.; Doehner, W.; von Haehling, S.; et al. Megestrol Acetate Improves Cardiac Function in a Model of Cancer Cachexia-Induced Cardiomyopathy by Autophagic Modulation. J. Cachexia Sarcopenia Muscle 2016, 7, 555–566. [Google Scholar] [CrossRef] [Green Version]
  72. Mishra, S.; Tamta, A.K.; Sarikhani, M.; Desingu, P.A.; Kizkekra, S.M.; Pandit, A.S.; Kumar, S.; Khan, D.; Raghavan, S.C.; Sundaresan, N.R. Subcutaneous Ehrlich Ascites Carcinoma Mice Model for Studying Cancer-Induced Cardiomyopathy. Sci. Rep. 2018, 8, 5599. [Google Scholar] [CrossRef] [PubMed]
  73. Mühlfeld, C.; Das, S.K.; Heinzel, F.R.; Schmidt, A.; Post, H.; Schauer, S.; Papadakis, T.; Kummer, W.; Hoefler, G. Cancer Induces Cardiomyocyte Remodeling and Hypoinnervation in the Left Ventricle of the Mouse Heart. PLoS ONE 2011, 6, e20424. [Google Scholar] [CrossRef] [PubMed]
  74. Law, M.L.; Metzger, J.M. Cardiac Myocyte Intrinsic Contractility and Calcium Handling Deficits Underlie Heart Organ Dysfunction in Murine Cancer Cachexia. Sci. Rep. 2021, 11, 23627. [Google Scholar] [CrossRef] [PubMed]
  75. Drott, C.; Waldenström, A.; Lundholm, K. Cardiac Sensitivity and Responsiveness to β-Adrenergic Stimulation in Experimental Cancer and Undernutrition. J. Mol. Cell. Cardiol. 1987, 19, 675–683. [Google Scholar] [CrossRef]
  76. Drott, C.; Ekman, L.; Holm, S.; Waldenström, A.; Lundholm, K. Effects of Tumor-Load and Malnutrition on Myocardial Function in the Isolated Working Rat Heart. J. Mol. Cell. Cardiol. 1986, 18, 1165–1176. [Google Scholar] [CrossRef]
  77. Liao, R.; Podesser, B.K.; Lim, C.C. The Continuing Evolution of the Langendorff and Ejecting Murine Heart: New Advances in Cardiac Phenotyping. Am. J. Physiol. Heart Circ. Physiol. 2012, 303, 156–167. [Google Scholar] [CrossRef]
  78. Burch, G.E.; Phillips, J.H.; Ansari, A. The Cachectic Heart. Dis. Chest 1968, 54, 403–409. [Google Scholar] [CrossRef]
  79. Sabbah, H.N. Silent Disease Progression in Clinically Stable Heart Failure. Eur. J. Heart Fail. 2017, 19, 469–478. [Google Scholar] [CrossRef] [Green Version]
  80. Goodpaster, B.H.; Carlson, C.L.; Visser, M.; Kelley, D.E.; Scherzinger, A.; Harris, T.B.; Stamm, E.; Newman, A.B. Attenuation of Skeletal Muscle and Strength in the Elderly: The Health ABC Study. J. Appl. Physiol. 2001, 90, 2157–2165. [Google Scholar] [CrossRef]
  81. Zhou, X.; Wang, J.L.; Lu, J.; Song, Y.; Kwak, K.S.; Jiao, Q.; Rosenfeld, R.; Chen, Q.; Boone, T.; Simonet, W.S.; et al. Reversal of Cancer Cachexia and Muscle Wasting by ActRIIB Antagonism Leads to Prolonged Survival. Cell 2010, 142, 531–543. [Google Scholar] [CrossRef] [Green Version]
  82. Shum, A.M.Y.; Poljak, A.; Bentley, N.L.; Turner, N.; Tan, T.C.; Polly, P. Proteomic Profiling of Skeletal and Cardiac Muscle in Cancer Cachexia: Alterations in Sarcomeric and Mitochondrial Protein Expression. Oncotarget 2018, 9, 22001–22022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Reed, A.L.; Tanaka, A.; Sorescu, D.; Liu, H.; Jeong, E.-M.; Sturdy, M.; Walp, E.R.; Dudley, S.C.; Sutliff, R.L. Diastolic Dysfunction Is Associated with Cardiac Fibrosis in the Senescence-Accelerated Mouse. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H824–H831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Cosper, P.F.; Leinwand, L.A. Cancer Causes Cardiac Atrophy and Autophagy in a Sexually Dimorphic Manner. Cancer Res. 2011, 71, 1710–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Lowes, B.D.; Gilbert, E.M.; Abraham, W.T.; Minobe, W.A.; Larrabee, P.; Ferguson, D.; Wolfel, E.E.; Lindenfeld, J.; Tsvetkova, T.; Robertson, A.D.; et al. Myocardial Gene Expression in Dilated Cardiomyopathy Treated with Beta-Blocking Agents. N. Engl. J. Med. 2002, 346, 1357–1365. [Google Scholar] [CrossRef]
  86. Miyata, S.; Minobe, W.; Bristow, M.R.; Leinwand, L.A. Myosin Heavy Chain Isoform Expression in the Failing and Nonfailing Human Heart. Circ. Res. 2000, 86, 386–390. [Google Scholar] [CrossRef] [Green Version]
  87. Herron, T.J.; Vandenboom, R.; Fomicheva, E.; Mundada, L.; Edwards, T.; Metzger, J.M. Calcium-Independent Negative Inotropy by β-Myosin Heavy Chain Gene Transfer in Cardiac Myocytes. Circ. Res. 2007, 100, 1182–1190. [Google Scholar] [CrossRef] [Green Version]
  88. Herron, T.J.; Devaney, E.; Mundada, L.; Arden, E.; Day, S.; Guerrero-Serna, G.; Turner, I.; Westfall, M.; Metzger, J.M. Ca2+ -independent Positive Molecular Inotropy for Failing Rabbit and Human Cardiac Muscle by A-myosin Motor Gene Transfer. FASEB J. 2010, 24, 415–424. [Google Scholar] [CrossRef] [Green Version]
  89. Milan, G.; Romanello, V.; Pescatore, F.; Armani, A.; Paik, J.-H.; Frasson, L.; Seydel, A.; Zhao, J.; Abraham, R.; Goldberg, A.L.; et al. Regulation of Autophagy and the Ubiquitin–Proteasome System by the FoxO Transcriptional Network during Muscle Atrophy. Nat. Commun. 2015, 6, 6670. [Google Scholar] [CrossRef] [Green Version]
  90. Zhang, Y.; Wang, J.; Wang, X.; Gao, T.; Tian, H.; Zhou, D.; Zhang, L.; Li, G.; Wang, X. The Autophagic-Lysosomal and Ubiquitin Proteasome Systems Are Simultaneously Activated in the Skeletal Muscle of Gastric Cancer Patients with Cachexia. Am. J. Clin. Nutr. 2020, 111, 570–579. [Google Scholar] [CrossRef]
  91. Springer, J.; Palus, S.; Rauchhaus, M.; Anker, S.D. Experimental Cancer Cachexia Severely Impairs Heart Function. J. Card. Fail. 2008, 14, S18. [Google Scholar] [CrossRef]
  92. Springer, J.; Tschirner, A.; Grzesiak, A.; Kaschina, E.; von Haehling, S.; Anker, S.D. Cancer Cachexia Therapy: A Matter of Helping the Heart? J. Card. Fail. 2010, 16, S12. [Google Scholar] [CrossRef]
  93. Manne, N.D.P.K.; Lima, M.; Enos, R.T.; Wehner, P.; Carson, J.A.; Blough, E. Altered Cardiac Muscle MTOR Regulation during the Progression of Cancer Cachexia in the ApcMin/+ Mouse. Int. J. Oncol. 2013, 42, 2134–2140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Hyltander, A.; Drott, C.; Körner, U.; Sandström, R.; Lundholm, K. Elevated Energy Expenditure in Cancer Patients with Solid Tumours. Eur. J. Cancer Clin. Oncol. 1991, 27, 9–15. [Google Scholar] [CrossRef]
  95. Wang, Z.; Ying, Z.; Bosy-Westphal, A.; Zhang, J.; Schautz, B.; Later, W.; Heymsfield, S.B.; Müller, M.J. Specific Metabolic Rates of Major Organs and Tissues across Adulthood: Evaluation by Mechanistic Model of Resting Energy Expenditure. Am. J. Clin. Nutr. 2010, 92, 1369–1377. [Google Scholar] [CrossRef]
  96. Weiss, R.G.; Gerstenblith, G.; Bottomley, P.A. ATP Flux through Creatine Kinase in the Normal, Stressed, and Failing Human Heart. Proc. Natl. Acad. Sci. USA 2005, 102, 808–813. [Google Scholar] [CrossRef] [Green Version]
  97. Gibbs, C.L. Cardiac Energetics. Physiol. Rev. 1978, 58, 174–254. [Google Scholar] [CrossRef]
  98. Aon, M.A.; Cortassa, S.; O’Rourke, B. Redox-Optimized ROS Balance: A Unifying Hypothesis. Biochim. Et Biophys. Acta (BBA) Bioenerg. 2010, 1797, 865–877. [Google Scholar] [CrossRef] [Green Version]
  99. Wisneski, J.A.; Gertz, E.W.; Neese, R.A.; Mayr, M. Myocardial Metabolism of Free Fatty Acids. Studies with 14C-Labeled Substrates in Humans. J. Clin. Investig. 1987, 79, 359–366. [Google Scholar] [CrossRef]
  100. Drott, C.; Lundholm, K. Glucose Uptake and Amino Acid Metabolism in Perfused Hearts from Tumor-Bearing Rats. J. Surg. Res. 1990, 49, 62–68. [Google Scholar] [CrossRef]
  101. Lee, D.E.; Brown, J.L.; Rosa-Caldwell, M.E.; Perry, R.A.; Brown, L.A.; Haynie, W.S.; Washington, T.A.; Wiggs, M.P.; Rajaram, N.; Greene, N.P. Cancer-induced Cardiac Atrophy Adversely Affects Myocardial Redox State and Mitochondrial Oxidative Characteristics. JCSM Rapid Commun. 2021, 4, 3–15. [Google Scholar] [CrossRef]
  102. Devine, R.D.; Bicer, S.; Reiser, P.J.; Wold, L.E. Increased Hypoxia-Inducible Factor-1α in Striated Muscle of Tumor-Bearing Mice. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H1154–H1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Tanaka, M.; Sugimoto, K.; Fujimoto, T.; Xie, K.; Takahashi, T.; Akasaka, H.; Kurinami, H.; Yasunobe, Y.; Matsumoto, T.; Fujino, H.; et al. Preventive Effects of Low-Intensity Exercise on Cancer Cachexia-Induced Muscle Atrophy. FASEB J. 2019, 33, 7852–7862. [Google Scholar] [CrossRef] [PubMed]
  104. Asp, M.L.; Tian, M.; Wendel, A.A.; Belury, M.A. Evidence for the Contribution of Insulin Resistance to the Development of Cachexia in Tumor-Bearing Mice. Int. J. Cancer 2010, 126, 756–763. [Google Scholar] [CrossRef]
  105. Copeland, G.P.; Leinster, S.J.; Davis, J.C.; Hipkin, L.J. Insulin Resistance in Patients with Colorectal Cancer. Br. J. Surg. 2005, 74, 1031–1035. [Google Scholar] [CrossRef] [PubMed]
  106. Raun, S.H.; Knudsen, J.R.; Han, X.; Jensen, T.E.; Sylow, L. Cancer Causes Dysfunctional Insulin Signaling and Glucose Transport in a Muscle-type-specific Manner. FASEB J. 2022, 36, e22211. [Google Scholar] [CrossRef] [PubMed]
  107. Pin, F.; Novinger, L.J.; Huot, J.R.; Harris, R.A.; Couch, M.E.; O’Connell, T.M.; Bonetto, A. PDK4 Drives Metabolic Alterations and Muscle Atrophy in Cancer Cachexia. FASEB J. 2019, 33, 7778–7790. [Google Scholar] [CrossRef] [Green Version]
  108. Fukawa, T.; Yan-Jiang, B.C.; Min-Wen, J.C.; Jun-Hao, E.T.; Huang, D.; Qian, C.-N.; Ong, P.; Li, Z.; Chen, S.; Mak, S.Y.; et al. Excessive Fatty Acid Oxidation Induces Muscle Atrophy in Cancer Cachexia. Nat. Med. 2016, 22, 666–671. [Google Scholar] [CrossRef]
  109. Wyart, E.; Reano, S.; Hsu, M.Y.; Longo, D.L.; Li, M.; Hirsch, E.; Filigheddu, N.; Ghigo, A.; Riganti, C.; Porporato, P.E. Metabolic Alterations in a Slow-Paced Model of Pancreatic Cancer-Induced Wasting. Oxidative Med. Cell. Longev. 2018, 2018, 6419805. [Google Scholar] [CrossRef] [Green Version]
  110. Thackeray, J.T.; Pietzsch, S.; Stapel, B.; Ricke-Hoch, M.; Lee, C.W.; Bankstahl, J.P.; Scherr, M.; Heineke, J.; Scharf, G.; Haghikia, A.; et al. Insulin Supplementation Attenuates Cancer-Induced Cardiomyopathy and Slows Tumor Disease Progression. JCI Insight 2017, 2, e93098. [Google Scholar] [CrossRef] [Green Version]
  111. Sassoon, D.A. Fatty Acid Metabolism-the First Trigger for Cachexia? Nat. Med. 2016, 22, 584–585. [Google Scholar] [CrossRef]
  112. Kühlbrandt, W. Structure and Function of Mitochondrial Membrane Protein Complexes. BMC Biol. 2015, 13, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Davies, K.M.; Anselmi, C.; Wittig, I.; Faraldo-Gómez, J.D.; Kühlbrandt, W. Structure of the Yeast F 1F O-ATP Synthase Dimer and Its Role in Shaping the Mitochondrial Cristae. Proc. Natl. Acad. Sci. USA 2012, 109, 13602–13607. [Google Scholar] [CrossRef] [Green Version]
  114. Hackenbrock, C.R. Ultrastructural Bases for Metabolically Linked Mechanical Activity in Mitochondria. I. Reversible Ultrastructural Changes with Change in Metabolic Steady State in Isolated Liver Mitochondria. J. Cell Biol 1966, 30, 269–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Brown, J.L.; Lawrence, M.M.; Borowik, A.; Oliver, L.; Peelor, F.F.; van Remmen, H.; Miller, B.F. Tumor Burden Negatively Impacts Protein Turnover as a Proteostatic Process in Noncancerous Liver, Heart, and Muscle, but Not Brain. J. Appl. Physiol. 2021, 131, 72–82. [Google Scholar] [CrossRef] [PubMed]
  116. Nukaga, S.; Mori, T.; Miyagawa, Y.; Fujiwara-Tani, R.; Sasaki, T.; Fujii, K.; Mori, S.; Goto, K.; Kishi, S.; Nakashima, C.; et al. Combined Administration of Lauric Acid and Glucose Improved Cancer-Derived Cardiac Atrophy in a Mouse Cachexia Model. Cancer Sci. 2020, 111, 4605–4615. [Google Scholar] [CrossRef] [PubMed]
  117. Powers, S.K.; Wiggs, M.P.; Duarte, J.A.; Murat Zergeroglu, A.; Demirel, H.A. Mitochondrial Signaling Contributes to Disuse Muscle Atrophy. Am. J. Physiol. Endocrinol. Metab. 2012, 303, E31–E39. [Google Scholar] [CrossRef]
  118. Yu, L.; Chen, Y.; Tooze, S.A. Autophagy Pathway: Cellular and Molecular Mechanisms. Autophagy 2018, 14, 207–215. [Google Scholar] [CrossRef] [Green Version]
  119. Melser, S.; Lavie, J.; Bénard, G. Mitochondrial Degradation and Energy Metabolism. Biochim. Et Biophys. Acta Mol. Cell Res. 2015, 1853, 2812–2821. [Google Scholar] [CrossRef] [Green Version]
  120. Mizushima, N.; Yoshimori, T. How to Interpret LC3 Immunoblotting. Autophagy 2007, 3, 542–545. [Google Scholar] [CrossRef]
  121. Pietzsch, S.; Ricke-Hoch, M.; Stapel, B.; Hilfiker-Kleiner, D. Modulation of Cardiac AKT and STAT3 Signalling in Preclinical Cancer Models and Their Impact on the Heart. Biochim. Et Biophys. Acta Mol. Cell Res. 2020, 1867, 118519. [Google Scholar] [CrossRef]
  122. Michaelis, K.A.; Zhu, X.; Burfeind, K.G.; Krasnow, S.M.; Levasseur, P.R.; Morgan, T.K.; Marks, D.L. Establishment and Characterization of a Novel Murine Model of Pancreatic Cancer Cachexia. J. Cachexia Sarcopenia Muscle 2017, 8, 824–838. [Google Scholar] [CrossRef] [PubMed]
  123. Clark, Y.Y.; Wold, L.E.; Szalacha, L.A.; McCarthy, D.O. Ubiquinol Reduces Muscle Wasting but Not Fatigue in Tumor-Bearing Mice. Biol. Res. Nurs. 2015, 17, 321–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Chance, B.; Williams, G.R. The Respiratory Chain and Oxidative Phosphorylation. Adv. Enzymol. Relat. Subj. Biochem. 2006, 17, 65–134. [Google Scholar]
  125. Gnaiger, E. Mitochondrial Pathways and Respiratory Control: An Introduction to OXPHOS. 5th ed. Bioenerg. Commun. 2020, 2020, 112. [Google Scholar]
  126. Schmidt, C.A.; Fisher-Wellman, K.H.; Darrell Neufer, P. From OCR and ECAR to Energy: Perspectives on the Design and Interpretation of Bioenergetics Studies. J. Biol. Chem. 2021, 297, 101140. [Google Scholar] [CrossRef]
  127. Abrigo, J.; Elorza, A.A.; Riedel, C.A.; Vilos, C.; Simon, F.; Cabrera, D.; Estrada, L.; Cabello-Verrugio, C. Role of Oxidative Stress as Key Regulator of Muscle Wasting during Cachexia. Oxidative Med. Cell. Longev. 2018, 2018, 2063179. [Google Scholar] [CrossRef]
  128. de Castro, G.S.; Simoes, E.; Lima, J.D.C.C.; Ortiz-Silva, M.; Festuccia, W.T.; Tokeshi, F.; Alcântara, P.S.; Otoch, J.P.; Coletti, D.; Seelaender, M. Human Cachexia Induces Changes in Mitochondria, Autophagy and Apoptosis in the Skeletal Muscle. Cancers 2019, 11, 1264. [Google Scholar] [CrossRef] [Green Version]
  129. Katerji, M.; Filippova, M.; Duerksen-Hughes, P. Approaches and Methods to Measure Oxidative Stress in Clinical Samples: Research Applications in the Cancer Field. Oxidative Med. Cell. Longev. 2019, 2019, 1279250. [Google Scholar] [CrossRef] [Green Version]
  130. Frijhoff, J.; Winyard, P.G.; Zarkovic, N.; Davies, S.S.; Stocker, R.; Cheng, D.; Knight, A.R.; Taylor, E.L.; Oettrich, J.; Ruskovska, T.; et al. Clinical Relevance of Biomarkers of Oxidative Stress. Antioxid. Redox Signal. 2015, 23, 1144–1170. [Google Scholar] [CrossRef] [Green Version]
  131. Hinch, E.C.A.; Sullivan-Gunn, M.J.; Vaughan, V.C.; McGlynn, M.A.; Lewandowski, P.A. Disruption of Pro-Oxidant and Antioxidant Systems with Elevated Expression of the Ubiquitin Proteosome System in the Cachectic Heart Muscle of Nude Mice. J. Cachexia Sarcopenia Muscle 2013, 4, 287–293. [Google Scholar] [CrossRef] [Green Version]
  132. Miyagawa, Y.; Nukaga, S.; Mori, T.; Fujiwara-Tani, R.; Fujii, K.; Mori, S.; Goto, K.; Kishi, S.; Sasaki, T.; Nakashima, C.; et al. Evaluation of Cancer-Derived Myocardial Impairments Using a Mouse Model. Oncotarget 2020, 11, 3712–3722. [Google Scholar] [CrossRef] [PubMed]
  133. Borges, F.H.; Marinello, P.C.; Cecchini, A.L.; Blegniski, F.P.; Guarnier, F.A.; Cecchini, R. Oxidative and Proteolytic Profiles of the Right and Left Heart in a Model of Cancer-Induced Cardiac Cachexia. Pathophysiology 2014, 21, 257–265. [Google Scholar] [CrossRef] [PubMed]
  134. Huot, J.R.; Pin, F.; Narasimhan, A.; Novinger, L.J.; Keith, A.S.; Zimmers, T.A.; Willis, M.S.; Bonetto, A. ACVR2B Antagonism as a Countermeasure to Multi-Organ Perturbations in Metastatic Colorectal Cancer Cachexia. J. Cachexia Sarcopenia Muscle 2020, 11, 1779–1798. [Google Scholar] [CrossRef] [PubMed]
  135. Powers, S.K.; Smuder, A.J.; Judge, A.R. Oxidative Stress and Disuse Muscle Atrophy: Cause or Consequence? Curr. Opin. Clin. Nutr. Metab. Care 2012, 15, 240–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Gülçin, I. Antioxidant Properties of Resveratrol: A Structure-Activity Insight. Innov. Food Sci. Emerg. Technol. 2010, 11, 210–218. [Google Scholar] [CrossRef]
  137. Shadfar, S.; Couch, M.E.; McKinney, K.A.; Weinstein, L.J.; Yin, X.; Rodriguez, J.E.; Guttridge, D.C.; Willis, M. Oral Resveratrol Therapy Inhibits Cancer-Induced Skeletal Muscle and Cardiac Atrophy in Vivo. Nutr. Cancer 2011, 63, 749–762. [Google Scholar] [CrossRef] [Green Version]
  138. Springer, J.; Tschirner, A.; Hartman, K.; von Haehling, S.; Anker, S.D.; Doehner, W. The Xanthine Oxidase Inhibitor Oxypurinol Reduces Cancer Cachexia-Induced Cardiomyopathy. Int. J. Cardiol. 2013, 168, 3527–3531. [Google Scholar] [CrossRef]
  139. Zhang, V.X.; Sze, K.M.F.; Chan, L.K.; Ho, D.W.H.; Tsui, Y.M.; Chiu, Y.T.; Lee, E.; Husain, A.; Huang, H.; Tian, L.; et al. Antioxidant Supplements Promote Tumor Formation and Growth and Confer Drug Resistance in Hepatocellular Carcinoma by Reducing Intracellular ROS and Induction of TMBIM1. Cell Biosci. 2021, 11, 217. [Google Scholar] [CrossRef]
  140. Assi, M.; Derbré, F.; Lefeuvre-Orfila, L.; Rébillard, A. Antioxidant Supplementation Accelerates Cachexia Development by Promoting Tumor Growth in C26 Tumor-Bearing Mice. Free Radic. Biol. Med. 2016, 91, 204–214. [Google Scholar] [CrossRef]
  141. Sayin, V.I.; Ibrahim, M.X.; Larsson, E.; Nilsson, J.A.; Lindahl, P.; Bergo, M.O. Cancer: Antioxidants Accelerate Lung Cancer Progression in Mice. Sci. Transl. Med. 2014, 6, 221ra15. [Google Scholar] [CrossRef]
  142. Wang, X.; Pickrell, A.M.; Zimmers, T.A.; Moraes, C.T. Increase in Muscle Mitochondrial Biogenesis Does Not Prevent Muscle Loss but Increased Tumor Size in a Mouse Model of Acute Cancer-Induced Cachexia. PLoS ONE 2012, 7, e33426. [Google Scholar] [CrossRef] [PubMed]
  143. Martinez, H.R.; Beasley, G.S.; Miller, N.; Goldberg, J.F.; Jefferies, J.L. Clinical Insights Into Heritable Cardiomyopathies. Front. Genet. 2021, 12, 663450. [Google Scholar] [CrossRef] [PubMed]
  144. Law, M.L.; Cohen, H.; Martin, A.A.; Angulski, A.B.B.; Metzger, J.M. Dysregulation of Calcium Handling in Duchenne Muscular Dystrophy-Associated Dilated Cardiomyopathy: Mechanisms and Experimental Therapeutic Strategies. J. Clin. Med. 2020, 9, 520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Howe, K.; Ross, J.M.; Loiselle, D.S.; Han, J.C.; Crossman, D.J. Right-Sided Heart Failure Is Also Associated with Transverse Tubule Remodeling in the Left Ventricle. Am. J. Physiol. Heart Circ. Physiol. 2021, 321, H940–H947. [Google Scholar] [CrossRef] [PubMed]
  146. Penna, F.; Busquets, S.; Argilés, J.M. Experimental Cancer Cachexia: Evolving Strategies for Getting Closer to the Human Scenario. Semin. Cell Dev. Biol. 2016, 54, 20–27. [Google Scholar] [CrossRef] [PubMed]
  147. Ballarò, R.; Costelli, P.; Penna, F. Animal Models for Cancer Cachexia. Curr. Opin. Supportive Palliat. Care 2016, 10, 281–287. [Google Scholar] [CrossRef] [PubMed]
  148. Herron, T.J.; Devaney, E.J.; Metzger, J.M. Modulation of Cardiac Performance by Motor Protein Gene Transfer. Ann. N. Y. Acad. Sci. 2008, 1123, 96–104. [Google Scholar] [CrossRef] [Green Version]
  149. Wessels, A.; Sedmera, D. Developmental Anatomy of the Heart: A Tale of Mice and Man. Physiol. Genom. 2004, 15, 165–176. [Google Scholar] [CrossRef]
Figure 1. Interrelationship between cancer cachexia and cardiac insufficiency is found in multiple common symptoms and mechanistic contributors. SNS (sympathetic nervous system).
Figure 1. Interrelationship between cancer cachexia and cardiac insufficiency is found in multiple common symptoms and mechanistic contributors. SNS (sympathetic nervous system).
Cells 11 01931 g001
Figure 2. Summary of cardiac mitochondrial changes in cancer cachexia that may contribute to impaired cardiac function.
Figure 2. Summary of cardiac mitochondrial changes in cancer cachexia that may contribute to impaired cardiac function.
Cells 11 01931 g002
Figure 3. Overview of cardiac alterations in cancer-induced cachexia. Both structural and metabolic remodeling likely contribute to decreased function.
Figure 3. Overview of cardiac alterations in cancer-induced cachexia. Both structural and metabolic remodeling likely contribute to decreased function.
Cells 11 01931 g003
Table 1. Overview of Clinical Studies Related to Cardiac Alterations in Cancer and Cancer-Induced Cachexia.
Table 1. Overview of Clinical Studies Related to Cardiac Alterations in Cancer and Cancer-Induced Cachexia.
Patients (Sample Size)Study DesignMajor Findings
Functional Remodeling
Anker, Ebner, Hildebrandt, et al. 2016 [45]NSCLC, pancreatic, CRC (145)Prospective, longitudinalHR > 75 bpm predicted for mortality
Anker, von Haehling, Coats, et al. 2021 [53]NSCLC, pancreatic, CRC (120)Prospective, longitudinal- ↑ NSVT vs. control
- NSVT & PVC predicted for mortality
Cramer, Hildebrandt, Kung, et al. 2014 [46]CRC (50)Prospective, single timepoint ↑ HR, ↓ HRV, ↓ LVEF vs. control
Kazemi-Bajestani, Becher, Butts, et al. 2019 [54]NSCLC (50)Prospective, longitudinal↓ LVEF, ↓ GLS at 4-month follow-up
Kazemi-Bajestani, Becher, Butts, et al. 2019 [56]NSCLC (70)Prospective, single timepoint↓ LVEF (<50%) incidence is higher in cachectic vs. non-cachectic patients
Lee, Park, Lim, et al. 2016 [44]Breast cancer (4786)Retrospective, longitudinal↑ HR predicted for mortality
Structural Remodeling
Barkhudaryan, Scherbakov, Springer, et al. 2017 [57]Lung, pancreatic, GI cancer (177)Retrospective, single timepoint↓ Heart weight in cachectic vs. non-cachectic patients
Cai, Mao, Yang, et al. 2020 [58]Pancreatic cancer (98)Retrospective, longitudinal↓ LVMA, LVMRA associated with mortality
Kazemi-Bajestani, Becher, Butts, et al. 2019 [54]NSCLC (50)Prospective, longitudinalCardiac atrophy associated with ↑ DLT, ↓ treatment response, ↓ physical functioning, ↑ mortality
Springer, Tschirner, Haghikia, et al. 2014 [48]NSCLC, pancreatic, GI cancer (12 cancer, 14 cancer cachexia)Post-mortemCachexia associated with ↓ heart mass, ↓ LVWT
Metabolic Remodeling
Hyltander, Körner, Lundholm, 1993 [59]Weight-losing cancer patients (60)Randomized, controlled trialSNS activation is a main driver of increased REE
Hyltander, Daneryd, Sandström, et al. 2000 [47]Weight-losing cancer patients (10)Randomized, crossoverSNS attenuation via β-blockers caused ↓ REE, ↓ HR, ↓ O2 uptake
Heckmann, Totakhel, Finke, et al. 2019 [60]Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, non-lymphatic cancer (337)Retrospective, single timepoint- Hodgkin’s lymphoma associated with ↑ cardiac glucose uptake
- Chemotherapy associated with ↓ cardiac glucose uptake
Colorectal cancer (CRC); dose-limiting toxicity (DLT); global longitudinal strain (GLS), heart rate (HR); heart rate variability (HRV); left ventricular ejection fraction (LVEF); left ventricular muscle area (LVMA); left ventricular muscle radiation attenuation (LVMRA); left ventricle wall thickness (LVWT); non-small cell lung cancer (NSCLC); non-sustained ventricular tachycardia (NSVT); premature ventricular contractions (PVC); resting energy expenditure (REE); sympathetic nervous system (SNS).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wiggs, M.P.; Beaudry, A.G.; Law, M.L. Cardiac Remodeling in Cancer-Induced Cachexia: Functional, Structural, and Metabolic Contributors. Cells 2022, 11, 1931. https://doi.org/10.3390/cells11121931

AMA Style

Wiggs MP, Beaudry AG, Law ML. Cardiac Remodeling in Cancer-Induced Cachexia: Functional, Structural, and Metabolic Contributors. Cells. 2022; 11(12):1931. https://doi.org/10.3390/cells11121931

Chicago/Turabian Style

Wiggs, Michael P., Anna G. Beaudry, and Michelle L. Law. 2022. "Cardiac Remodeling in Cancer-Induced Cachexia: Functional, Structural, and Metabolic Contributors" Cells 11, no. 12: 1931. https://doi.org/10.3390/cells11121931

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