Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Aug 19.
Published in final edited form as: JACC Heart Fail. 2024 Apr 8;12(5):795–809. doi: 10.1016/j.jchf.2024.02.021

Aging in Heart Failure

Embracing Biology Over Chronology: JACC Family Series

Parag Goyal a, Mathew S Maurer b, Jason Roh c
PMCID: PMC11331491  NIHMSID: NIHMS1991920  PMID: 38597865

Abstract

Age is among the most potent risk factors for developing heart failure and is strongly associated with adverse outcomes. As the global population continues to age and the prevalence of heart failure rises, understanding the role of aging in the development and progression of this chronic disease is essential. Although chronologic age is on a fixed course, biological aging is more variable and potentially modifiable in patients with heart failure. This review describes the current knowledge on mechanisms of biological aging that contribute to the pathogenesis of heart failure. The discussion focuses on 3 hallmarks of aging—impaired proteostasis, mitochondrial dysfunction, and deregulated nutrient sensing—that are currently being targeted in therapeutic development for older adults with heart failure. In assessing existing and emerging therapeutic strategies, the review also enumerates the importance of incorporating geriatric conditions into the management of older adults with heart failure and in ongoing clinical trials.

Keywords: aging biology, geriatrics, heart failure


Heart failure (HF), a heterogenous clinical syndrome of pulmonary or systemic congestion caused by a structural and/or functional abnormality in the heart, disproportionately affects older adults.1 Age is among the most potent risk factors independently associated with HF, regardless of ejection fraction, and its outcomes. Although the passage of time allows for greater exposure to known causes of HF (eg, coronary artery disease), even in the absence of overt myocardial injury, the aging cardiovascular system mirrors many of the phenotypes in HF2,3 (Central Illustration).4 This substrate places older adults at particularly high risk of developing HF, where even minor perturbations can shift this vulnerable homeostatic state toward the pathologic state of HF. Indeed, the risk of developing HF is ~20-fold higher among adults ≥60 years old, compared with their younger counterparts.5

CENTRAL ILLUSTRATION. Aging Processes in Heart Failure.

CENTRAL ILLUSTRATION

Physiological and biological aging of the cardiovascular system underlie the strong association between chronologic age and heart failure. This figure depicts the evolution of how “age” is being studied in heart failure and the shifting importance from chronologic to biological age in management of older adults with heart failure. The figure incorporates the updated hallmarks of aging outlined by Lopez-Otin et al9 and was partly generated using Servier Medical Art, provided by Servier, licensed under a Creative Commons Attribution 3.0 unported license.

Although the association between chronologic age and HF risk is well established, our understanding of the mechanisms underlying this relationship remains incomplete, and this has limited therapeutic development in HF. This issue has implications on advancing care for the rapidly growing older adult population with HF, which notably includes an increasing number of septagenarians, octagenarians, and nonagenarians in the United States and beyond. In response to this knowledge gap, the National Institutes of Health launched a Geroscience Initiative in 2012 with the following goals: 1) elucidating how biological aging contributes to the pathogenesis of chronic diseases, such as HF, in older adults; and 2) harnessing this knowledge to develop new therapeutics to prevent or delay these diseases and slow the functional decline associated with age.6

Recent reviews have highlighted the emerging role of geroscience in the broader scope of cardiovascular disease (CVD).7,8 Here we review how geroscience is providing new insights into HF pathophysiology and leading to the development of novel gerotherapeutic strategies for HF. Additionally, we enumerate an approach to incorporating geriatric conditions into the management of older adults with HF as a window into systemic biological age and emphasize the need to integrate geriatric conditions into clinical trials to assess the impact of established and emerging therapeutics.

BIOLOGICAL AGING: AN EMERGING THERAPEUTIC TARGET FOR HF

The distinction between chronologic and biological age is a core principle in geriatric medicine. Chronologic age is easy to recognize (age in years following birth) and progresses at a predictable rate. Biological age is more variable and is assessed by various metrics, including telomere length, epigenetic clocks, and clinical frailty phenotypes. The processes governing biological age are hypothesized to be common drivers of chronic disease pathogenesis in older adults. In other words, whereas chronologic age is on a fixed path, biological age is potentially modifiable. Thus, intervening on biological age could have broad implications on how we treat chronic diseases such as HF.

In accordance with this central geroscience concept, Lopez-Otin et al4,9 published the first reports on the “hallmarks of aging,” where they described 12 fundamental biological processes that dictate organismal aging Although the study of aging is centuries old, this newer concept of “aging hallmarks” has provided a useful framework for investigating how mechanisms of biological aging contribute to HF (Central Illustration). Here we focus on 3 specific hallmarks of aging—impaired proteostasis, mitochondrial dysfunction, and deregulated nutrient sensing—in which emerging evidence supports a causal role in cardiac aging and HF and is leading to the translation of gerotherapeutics specifically in older adults with HF. It is important to emphasize that all 12 hallmarks of aging contribute to HF pathophysiology, and many of these hallmarks have been the topics of excellent reviews in cardiovascular aging.1016 Moreover, as highlighted by the Unitary Theory of Fundamental Aging Mechanisms, there are complex interactions and interdependence among these processes, whereby each hallmark can affect another, thereby making it unlikely that any single hallmark functions in isolation.17 Although reviewing all 12 hallmarks in depth is beyond the scope of this review, Table 1 provides an overview of recent and emerging clinical trials targeting the various hallmarks of aging in HF.

TABLE 1.

Heart Failure Trials Targeting Select Hallmarks of Aging Biology

Hallmark of Aging Therapeutic Interventions/Target Impact

Impaired proteostasis/ autophagy • Tafamidis (transthyretin [TTR] stabilizer)
• Acoramidis (TTR stabilizer)
• Patisiran (TTR silencer)
• NTLA-2001 (TTR gene editing)a
• ALXN-2220 (formerly NI006) (TTR clearance antibody)
• Spermidine (autophagy)a
• ↓ CV hospitalizations, ↓ mortality, slows decline in functional capacity and QoL (ATTR-CA)
• ↓ CV hospitalizations, ↓ all-cause mortality, attenuates rise in NT-proBNP, slows decline in 6MWT (ATTR-CA)
• Preserves functional capacity and QoL, stablized cardiac function (ATTR-CA)
NCT06183931 (ATTR-CA)
• ↓ ECV, ↓ cardiac tracer uptake on scintigraphy, ↓ NT-proBNP (ATTR-CA)
NCT05128331 (HFpEF)
Mitochondrial dysfunction • Coenzyme Q10
• Alpha lipoic acid
• Nicotinamide riboside (NAD+)a
• Elamipretide
• Perhexiline (CPT)
• PUFA
• Ketone estera
• ↓ CV hospitalizations, ↓ mortality, NC/↑ LVEF, NC/↑ 6MWT, NC/↑ NYHA (HFrEF); NC diastolic function, NC NT-proBNP (HFpEF)
• ↓ Troponin, ↓ inflammation, ↑ peak systolic strain (diabetic CMP)
• ↓ Inflammation, NCT04528004 (HFrEF)
• NC LVEF, NC LVESV (HFrEF); NCT02814097 (HFpEF)
• NC/↑ LVEF, ↑ NYHA, ↑ QoL, ↑ exercise capacity (HFrEF)
• ↓ CV hospitalizations, ↓ mortality, ↑ LVEF (HFrEF/HFpEF)
NCT05348460 (HFrEF)
Deregulated nutrient sensing • Rapamycin (mTOR)a
• Metformin (AMPK)a
• ↑ Physical function; NCT04996719 (HFpEF)
• NC LVEF, exercise capacity (HFrEF); NCT05093959 (HFpEF)
Inflammation • Canakinumab (IL-1β)
• Anakinra (IL-1)
• Colchicine (tubulin)a
• Methotrexate (multiple)
• Infliximab (TNFα)
• ↓ HHF, ↓ HF-related mortality (iCMP)
• ↓ NT-proBNP, ↑ exercise capacity (<4 wks), NC exercise capacity (>12 weeks), NC HHF, NC mortality (HFrEF and HFpEF)
• NC NYHA, NC HHF, NC mortality; NCT05637398 (HFpEF)
• NC HHF, NC 6MWT, NC QoL (iCMP)
• NC HHF, NC mortality; [ mortality (with higher doses) (HFrEF)
Altered intercellular communication • Pirfenidone (fibrosis)a
• Allogenic derived cells (fibrosis)a
NCT02932566 (HFpEF)
NCT02941705 (HFpEF)
Telomere shortening • AAV9-cTnT-modTERT (telomerase)a NCT05837143 (HFrEF)
Epigenetic alterations • CDR132L (miR-132 inhibitor) a
• TN-301 (HDAC6)
• ↓ NT-proBNP, NCT05350969 (HFrEF/iCMP); NCT05953831 (HFpEF) HFpEF
Cellular senescence • Fisetina NCT06133634, NCT03675724 (assessing vascular function, frailty, inflammation in older adults; HF not excluded)
Dysbiosis • Rifaxamin, Saccharomyces boulardiia
• Empagliflozin a
• Ensure Immunonutrition Shakea
Lactobacillus plantaruma
• Acacia gum
NCT02637167 (HFrEF)
NCT05584319 (HFpEF)
NCT05655910 (HFrEF)
NCT05752760 (HFpEF/HFrEF)
NCT03409926 (HFpEF/HFrEF)
Stem cell exhaustion • Mesenchymal stromal cells
• BM-MNC (with or without G-CSF)
• C-kit+ cardiac cells
• CD34+ cells
• NC/↓ HHF, NC/↓ cardiacdeath, NC/↑ QoL, ↑ 6MWT, NC/↓ LVEDV, NC/↑ LVEF, (HFrEF)
• NC/↑ LVEF, functional capacity, NYHA, QoL (HFrEF)
• ↓ HHF, NC exercise capacity, NC 6MWT, NC LVEF, NC LV volumes, NC NT-proBNP,
• ↓ Mortality, ↑ LVEF, NC LV volumes, ↑ 6MWT, ↓ NT-proBNP (HFrEF)
a

Active clinical trial.

6MWT = 6-minute walk time; AMPK = AMK-activated protein kinase; ATTR-CA = transthyretin cardiac amyloidosis; BM-MNC = bone marrow-derived mononuclear cells; CPT = carnitine palmitoyltransferase; CV = cardiovascular; ECV = extracellular volume; G-CSF = granulocyte colony stimulating factor; HDAC6 = histone deacetylase 6; HF = heart failure; HFpEF = heart failure with preserved ejection fraction; HFrEF = heart failure with reduced ejection fraction; HHF = hospitalizations for heart failure; iCMP = ischemic cardiomyopathy; IL = interleukin; LV = left ventricular; LVEDV = left ventricular end-diastolic volume; LVEF = left ventricular ejection fraction; LVESV = left ventricular end-systolic volume; mTOR = mechanistic target of rapamycin; NAD+ = nicotinamide adenine dinucleotide; NC = no change; NT-proBNP = N-terminal pro-B-type natriuretic peptide; PUFA = polyunsaturated fatty acid; QoL = quality of life; TNF = tumor necrosis factor.

IMPAIRED PROTEOSTASIS

BIOLOGY IN AGING AND DISEASE.

A universal hallmark of cellular aging is the decline in intracellular quality control systems. Proteostasis is the process by which cells regulate their protein content, which in humans consists of ~1 to 3 billion proteins per cell. When proteostasis is compromised, misfolded or aggregated proteins accumulate, leading to impairments in cellular function and survival. Given the greater impact of proteotoxicity in postmitotic cells, such as neurons and cardiomyocytes, the nervous and cardiovascular systems are disproportionately affected. Not surprisingly, loss of proteostasis contributes to the causal pathobiology in Alzheimer dementia18 and cardiac amyloidosis.19

Proteostasis is regulated by highly dynamic and complex intracellular machinery that encompasses molecular chaperones (eg, heat shock proteins) that facilitate proper folding and translocation of proteins, stress-responsive pathways such as the unfolded protein response (UPR), and proteolytic pathways such as the ubiquitin-proteasome (UPS) and autophagosome-lysosome systems. These components ultimately decide the fate of misfolded proteins and dictate whether they will be refolded or eliminated.

ROLE IN CARDIAC AGING AND HF.

With aging, impairments in all arms of the proteostasis network occur.20 The best evidence for a causal role of this biology in cardiac aging and HF comes from cardiac-specific manipulation of this process in animal models. Inhibition of autophagy, UPS, or UPR in the heart is sufficient to induce HF in young animals, whereas restoring these processes, through either cardiomyocyte-specific genetic manipulation or pharmacologic activators (eg, spermidine), can reverse adverse myocardial remodeling and cardiac dysfunction in animal models of cardiac aging or HF with preserved ejection fraction (HFpEF).2124 Moreover, forced cardiac overexpression of preamyloid oligomers or mutated chaperones is sufficient to induce cardiac hypertrophy and HF in animal models of cardiac amyloidosis or desmin-related cardiomyopathy.25,26 Age-associated defects in hepatic chaperone and UPR capacity also increase cardiac transthyretin (TTR) deposition,27 and inhibiting cardiomyocyte autophagy accelerates intracellular protein aggregation, myocardial fibrosis, and cardiac dysfunction in animal models of desmin-related cardiomyopathy.28 Taken together, these preclinical studies strongly implicate a causal role of impaired proteostasis in HF.

THERAPEUTIC DEVELOPMENT.

Targeting impaired proteostasis mechanisms in age-related HF syndromes has been a recent bench-to-bedside success story with pharmacologic agents now part of guideline-directed management for cardiac amyloidosis.29,30 TTR stabilizers have garnered regulatory approval on the basis of successful phase III trials in TTR amyloid cardiomyopathy, the prototypical disease of impaired proteostasis that almost exclusively affects older adults. Data from these trials demonstrate robust and consistent effects of stabilizing TTR proteins with tafamidis and acoramidis, which reduce cardiovascular and overall hospitalizations.30 More recently, RNA interference therapeutics that genetically inhibit TTR production in hepatocytes have shown that this approach can also stabilize this progressive and previously fatal form of HF.31 Phase III trials are also being initiated with an in vivo, systemically delivered, investigational CRISPR-based therapy designed to inactivate the TTR gene in liver cells and thereby prevent the production of TTR protein,32 whereas humanized monoclonal antibodies specifically targeting misfolded TTR have shown early evidence of efficacy in TTR cardiac amyloidosis.33

MITOCHONDRIAL DYSFUNCTION

BIOLOGY IN AGING AND DISEASE.

Mitochondria are central regulators of cellular metabolism and energetics. They not only serve as the primary site for adenosine triphosphate (ATP) generation, but in doing so they also underlie the free radical theory of aging, which proposes that oxidative stress is responsible for the cumulative cellular damage that causes aging.34 Although this theory has been somewhat controversial, oxidative stress from mitochondrial dysfunction is widely accepted as an important mechanism in age-related disorders, including dementia,35 frailty/sarcopenia,36 and HF.37

Multiple factors contribute to age-associated mitochondrial dysfunction, including self-perpetuating oxidative damage, nicotinamide adenine dinucleotide (NAD+) depletion, reduced mitogenesis, impaired mitophagy, morphologic changes, and somatic mutations.38,39 Mitochondria notably possess their own genome (mtDNA), and the accumulation of mtDNA mutations contributes to age-associated mitochondrial dysfunction. The PolG mutator mouse provides direct evidence for a causal role of mitochondrial dysfunction in aging. Defective mitochondrial DNA polymerase and ineffective mtDNA proofreading in these mice led to the accumulation of mtDNA mutations and oxidative stress and subsequently accelerated aging phenotypes in multiple organs, including the heart.38,40,41

ROLE IN CARDIAC AGING AND HF.

With its high energy demands, the heart possesses the highest content of mitochondria among all organs. Mitochondria constitute ~30% of the total volume in cardiomyocytes and are responsible for generating >90% of the ATP used by the heart. In the aged heart, deleterious changes in mitochondrial morphology, membrane integrity, volume, and function all occur.39,42 These changes are not only associated with increased reactive oxygen species (ROS)–mediated cellular damage and metabolic reprogramming (a shift from fatty acid [FA] oxidation to glycolytic metabolism), but they also intersect with other mechanisms of cardiac aging, including dysregulated calcium (Ca2+) cycling, proteostasis, metainflammation, and senescence.37,39,43 Definitive evidence for a causal role of mitochondrial dysfunction and oxidative stress in cardiac aging comes from mice in which mitochondrial catalase is overexpressed. These mice exhibit reduced cardiac ROS and attenuation of nearly all cardiac aging phenotypes.41,43 Gain- and loss-of-function data of peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1), a master regulator of mitochondrial biogenesis, also support an important role for mitochondrial dysfunction in cardiac aging and HF. Genetic deletion of PGC-1α in the heart accelerates the age-related decline in cardiac function and resilience to injury.44,45 Although forced overexpression of cardiac PGC-1α induces robust mitochondrial biogenesis, it requires careful titration because mitochondrial oversaturation in the heart can also amplify ROS production and induce cardiac failure.46,47

THERAPEUTIC DEVELOPMENT.

Despite strong evidence supporting a causal role of mitochondrial dysfunction and oxidative stress in cardiac aging and HF, randomized clinical trials of antioxidant interventions in HF have generally been disappointing.48 This result in part stems from strategies previously used, which mostly relied on antioxidant supplementation that may not have effectively modulated oxidative stress within the heart. More recently, coenzyme Q (CoQ), a free radical scavenging component of the electron transport chain, and lipoic acid, a cofactor for 2-ketoacid dehydrogenases, have shown promising results in animals and humans.49,50 CoQ10 declines with age and HF, and lifelong supplementation in rodents effectively reduces age-associated cardiac mitochondrial dysfunction and oxidative stress.51 Q-SYMBIO (Coenzyme Q10 as adjunctive treatment of chronic heart failure: a randomised, double-blind, multicentre trial with focus on SYMptoms, BIomarker status [Brain-Natriuretic Peptide (BNP)], and long-term Outcome [hospitalisations/mortality]), a randomized trial of CoQ10 treatment in HF, showed that 2 years of CoQ10 improved mortality, HF hospitalization, and NYHA functional class.52 Interestingly, trends toward stronger effects were observed in older adults and in those with higher left ventricular ejection fraction. A small pilot study of older adults with HFpEF did not show benefit of CoQ10 on diastolic function or N-terminal pro–B-type natriuretic peptide.53 However, treatment lasted only 4 months, and this trial was not powered to assess HF outcomes. Lipoic acid also improves cardiac mitochondrial function and redox homeostasis, and it can reverse toxic lipid accumulation and dysfunction in animal models of aging and HF.54,55 A small pilot study of diabetic cardiomyopathy showed that lipoic acid supplementation reduces peripheral biomarkers of oxidative stress and inflammation and improves subclinical cardiac dysfunction.56 A trial of patients with diabetes and ischemic cardiomyopathy is now underway to investigate the use of this antioxidant further in HF (Alpha-lipoic Acid in Diabetic Patients With Ischemic Cardiomyopathy; NCT06056687).

Recent work has suggested that directly targeting mitochondrial dysfunction may be more effective. NAD+ repletion is one such strategy being investigated (Mechanistic Studies of Nicotinamide Riboside in Human Heart Failure; NCT04528004). NAD+ is a critical coenzyme necessary for mitochondrial redox reactions, and its age-associated decline contributes to mitochondrial dysfunction and increased ROS. In animal models of HF, boosting NAD+ levels with its precursor nicotinamide riboside improves mitochondrial function, myocardial energetics, and cardiac function.57,58 Early data in clinical trials show that nicotinamide riboside safely increases NAD+ levels and potentially reduces inflammation in humans.59 Whether it improves HF outcomes remains to be determined. The mitochondrial targeted peptide, MTP-131 (elamipretide), is another candidate therapeutic being investigated in aging and HF. MTP-131 is a cell-permeable Szeto-Schiller peptide that uniquely concentrates in the inner mitochondrial membrane where it enhances ATP synthesis by inhibiting cardiolipin peroxidation. MTP-131 rapidly restores muscle mitochondrial energetics in older animals and humans,60,61 and it improves mitochondrial respiration and cardiac function in animal models of HF.62,63 Although MTP-131 did not improve left ventricular ejection fraction in a small study of patients with HF with reduced ejection fraction (HFrEF),64 its effects in HFpEF are still being investigated (Study to Evaluate the Effects of 4 Weeks Treatment With Subcutaneous Elamipretide on Left Ventricular Function in Subjects With Stable Heart Failure With Preserved Ejection Fraction; NCT02814097).

Finally, modulating the associated metabolic reprogramming that occurs in cardiac aging and HF is an alternative strategy to address mitochondrial dysfunction. Perhexiline, an antianginal drug that inhibits mitochondrial carnitine palmitoyltransferase (CPT)-1 and CPT-2, shifts muscle metabolism from FA to glucose use. In patients with HFrEF, perhexiline improved cardiac function, exercise capacity, and muscle energetics.65,66 Interestingly, despite inducing changes in cardiac and skeletal muscle energetics, perhexiline did not alter substrate use, a finding suggesting that its effects may be mediated by different mechanisms of action. Although no clinical trials are currently underway in older adults with HF, small pilot studies have shown symptomatic benefit in older adults with severe aortic stenosis.67 Reducing FA substrate availability to the heart may also attenuate cardiac lipotoxicity generated by a bottleneck effect from impaired mitochondrial FA oxidation in the aged and failing heart.68 Circulating levels of FAs (eg, triglycerides) increase with aging, and they can be effectively reduced with omega-3 and omega-6 polyunsaturated FAs (PUFAs). Higher circulating PUFAs have been associated with reduced HF risk in humans,69 and in mice, PUFA supplementation can prevent adverse myocardial remodeling and dysfunction induced by pressure overload.70 The GISSI-HF (Gruppo Italiano per lo Studio della Sopravivenza nell’Insufficienza Cardiaca–Heart Failure) trial, a double-blind, placebo-controlled randomized clinical trial of nearly 7,000 patients with HF, showed that PUFA therapy reduces mortality and cardiovascular hospitalizations.71 In older adult men at high risk of CVD, PUFA treatment also showed a trend toward reduced overall mortality, but no differences in cardiovascular events, although HF was not included in this outcome.72 Although PUFA supplementation is not specifically being tested in older adults with HF, the generally favorable safety profile and potential efficacy of this intervention may warrant future investigations. Indeed, the overwhelming beneficial effects of sodium glucose co-transporter-2 inhibitors and glucagon-like peptide 1 receptor agonists in HFrEF and HFpEF are likely partly the result of optimization of substrate use, thereby supporting this approach in HF therapeutic development.

DEREGULATED NUTRIENT SENSING

BIOLOGY IN AGING AND DISEASE.

Nutrient sensing pathways not only closely intersect with mitochondria biology, but are also some of the most conserved and powerful regulators of life span.4,9 An organism’s ability to respond to changes in nutritional states, from nutrient excess to scarcity, provides the metabolic adaptability and stress resilience necessary for optimal function and survival. Nutrients, such as glucose, FAs, amino acids, and ketones, are substrates for energy production and are primarily regulated by 4 major pathways: the insulin/insulin-like growth factor (IGF)-1, mechanistic target of rapamycin (mTOR), sirtuin, and AMK-activated protein kinase (AMPK) pathways. Insulin/IGF-1 and mTOR signaling activate under conditions of nutrient excess, generating an anabolic program in the body, whereas sirtuins and AMPK respond to low-energy states by detecting high levels of NAD+ or adenosine monophosphate. In general, the catabolic processes associated with the latter enhance life span, whereas chronic activation of IGF-1 and mTOR pathways tend to accelerate aging and functional decline.

ROLE IN CARDIAC AGING AND HF.

The role of nutrient-sensing pathways in the heart is complex, and their effects on cardiac function and remodeling are highly dependent on context, chronicity, and intensity of activation. For example, exercise transiently activates IGF-1/mTOR/Akt signaling, which is necessary for the physiological cardiac hypertrophy that mediates many of the functional and cardioprotective benefits of exercise.73,74 However, chronic activation of these anabolic pathways can also lead to unrestrained pathologic hypertrophy and cardiac failure.75 Similarly, although decreased IGF-1 signaling is associated with longevity, the age-dependent decline in circulating IGF-1 paradoxically correlates with increased HF risk in older adults.76 Recent work in cardiac-specific transgenic mice in which cardiac IGF-1 receptor signaling is chronically enhanced or reduced revealed that the effects of this nutrient-sensing pathway may be age dependent, improving cardiac function in young animals but conversely impairing cardiac autophagy, energetics, and function in old animals.77

In cardiac aging, mTOR is among the most extensively studied of the nutrient-sensing pathways. Its increased activity is associated with both age- and chronic stress-induced cardiac hypertrophy and dysfunction, impaired proteostasis/autophagy, and mitochondrial dysfunction.78 Although chronic activation of cardiac mTOR induces HF in animal models, the converse is not as straightforward. Complete inhibition of cardiac mTOR activity in adulthood similarly leads to HF by interfering with physiological cardiac growth necessary for survival. However, partial or transient mTOR inhibition with pharmacologic agents, such as rapamycin, exhibits striking beneficial effects in aged animals, by reversing many of the pathologic phenotypes associated with cardiac aging, along with enhancing longevity.7981

THERAPEUTIC DEVELOPMENT.

Interest in targeting nutrient-sensing pathways in aging largely stems from the robust and consistent effects of caloric restriction (CR) on longevity in nonhuman animal species. The primary mechanism by which CR enhances life span and health span is through modulation of nutrient-sensing pathways, activation of sirtuins and AMPK, and suppression of mTOR and IGF-1 signaling. In animal models of aging and HF, CR consistently attenuates age-associated cardiac dysfunction, adverse myocardial remodeling, oxidative stress, mitochondrial dysfunction, and loss of proteostasis.82 In humans, the effects of CR are not as clear, perhaps in part because these effects may be related to the consistency and intensity of CR regimens. Observational studies have suggested that CR is associated with longer life span, favorable cardiometabolic profiles, and reduced systemic inflammation.83 Although randomized trials have shown that CR interventions lasting 2 years have similar effects on cardiometabolic profiles and inflammation, no significant effects were detected on biological aging, as measured by DNA methylation epigenetic clocks.8385 Moreover, whereas CR has been shown to have modest effects on age-associated diastolic dysfunction in healthy older adults and improves exercise capacity in obese older adults with HFpEF, it did not alter cardiac structure or function in the latter group.86,87 Taken together, these data highlight both the complexity and potential of intervening on nutrient-sensing pathways.

Given the technical and logistical challenges of long-term CR, mimetics, such as rapamycin, are being explored as therapeutic agents for multiple age-associated chronic diseases, including HF. Although the immunosuppressive effects of mTOR inhibitors were an initial concern, clinical studies have shown that these agents actually improve immune function and reduce immunosenescence in older adults.88 A small pilot study of older adults with CVD enrolled in cardiac rehabilitation similarly showed that low-dose rapamycin was safe, decreased some senescence and inflammatory markers, and improved physical performance metrics, but not overall frailty.89 Early phase clinical trials are now underway to test the efficacy of rapamycin in older adults with HFpEF (Effect of Rapamycin to Improve Cardiac Function in Frail Older Adults; NCT04996719).

AMPK activation is also being explored as a potential therapeutic target, specifically through use of the common diabetes drug, metformin. Metformin is a synthetic biguanide that was first generated more than a century ago and was introduced as a treatment for diabetes in the 1950s. Interestingly, independent of its effects on diabetes, metformin reduces all-cause mortality and incidence of cancer and CVD.90 The mechanisms by which metformin enhances health span in older animals is multifold, but they largely stem from the drug’s effects on nutrient-sensing pathways, by suppressing both IGF and mTOR signaling while activating AMPK.91 In animal models of HF, the cardioprotective effects of metformin are thought to be primarily derived from AMPK activation, which has downstream effects on autophagy, oxidative stress, inflammation, mitochondrial function, and sirtuin activation.92,93 Current clinical trials are assessing the efficacy of metformin not only in preventing age-related chronic diseases, including HF,94 but also in treating HFpEF in older adults (Metformin for Older Patients With Heart Failure With Preserved Ejection Fraction; NCT05093959).

EXERCISE: A QUINTESSENTIAL GEROTHERAPEUTIC INTERVENTION

As highlighted throughout this review, the aforementioned hallmarks of aging do not function independently of each other. Rather, they intersect in a complex network at the cellular and organ level, which essentially dictates a variable and heterogeneous rate of aging in any given individual.95,96 Addressing this complexity is challenging but in many ways is achieved by exercise. Physical activity affects all hallmarks of aging and enhances resiliency in essentially every organ system.97 The aging cardiovascular system is no exception. Exercise not only attenuates or reverses many of the cardiac phenotypes associated with aging, but it also improves inflammation, mitochondrial function, proteostasis, metabolism, epigenetic alterations, telomerase activity, and even regenerative potential of the heart.3,98101 Notably, the loss of cardiomyocytes is central to the pathogenesis of HF, and addressing this key deficit has been considered one of the holy grails in the field. Although stem cell interventions in HF have been somewhat controversial, recent studies have shown that exercise, in itself, can enhance the declining regenerative potential of the aging cardiomyocyte.99,102 Thus, understanding the mechanisms by which exercise promotes cardiomyogenesis could help advance the fields of regenerative medicine and cell therapeutics in HF.

Given the pleiotropic effects of exercise on biological aging and its overwhelming beneficial impact in patients with HF, including older adults,87,103,104 this lifestyle intervention provides a valuable “gerotherapeutic” tool for not only improving outcomes in older adults with HF, but also gaining new mechanistic insights into the biology of cardiovascular aging. Synergistic efforts in preclinical models, human -omics (MoTrPAC [Molecular Transducers of Physical Activity Consortium]), and interventional clinical trials in older adults with HFpEF (Physical Rehabilitation for Older Patients With Acute Heart Failure With Preserved Ejection; NCT05525663) are underway to investigate the role of exercise in human aging, health, and disease.

BEYOND THE HEART: CONSIDERATION OF GERIATRIC CONDITIONS

A hallmark feature of organismal aging is the relentless, time-dependent decline in physiological reserves across all organ systems. Although each system does not “age” at the same rate, the progression of this process is inevitable in all organ systems, which are interconnected through a complex systemic biology.95,96 Thus, not only will changes in organ function occur throughout the body, but also the acceleration or slowing of this process in 1 organ can influence the aging and function of other organs. Coupled with interactions with lifestyle and environment, aging leads to widening in the variance of physiological parameters associated with age. This core principle of aging underlies the heterogeneity of phenotypes in older adults with HF (Figure 1).105

FIGURE 1. Interaction of Biological Aging With Environment and Lifestyle to Generate Heart Failure Phenotype.

FIGURE 1

Interactive concentric model (adapted from Inouye et al105) displaying how biological aging interacts with environment and lifestyle to cause geriatric conditions and heart failure (HF) in older adults. Identifying synergistic pathways offers a locus for the rational design and development of targeted interventions and gerotherapeutics

Geriatric conditions reflect this interdependence, representing manifestations of biological aging processes that span multiple organ systems. These conditions occur when the accumulated deficits in multiple systems render an older person vulnerable to situational challenges. Several geriatric conditions, including malnutrition and cachexia, sarcopenia, frailty, and cognitive impairment, coexist with HF in part because of shared mechanisms of dysregulated biological aging, including inflammation, impaired proteostasis, mitochondrial dysfunction, and deregulated nutrient sensing. Table 2 provides definitions of each geriatric condition, key aging processes shared with HF, and their estimated prevalence in HF.93,99103

TABLE 2.

Definition and Prevalence of Select Geriatric Conditions

Geriatric Condition Definition Key Aging Processes Shared With HF Prevalence in HF a

Malnutrition A deficit in protein-energy and micronutrients intake 37%-56%
Cachexia93,99 An extreme form of malnutrition and is defined as a systemic multifactorial catabolic circumstance characterized by generalized wasting of body compositions Inflammaging 10%-15%
Sarcopenia99,100 Progressive loss of muscle strength, mass, and function Consensus definitions differ:
• Per Sarcopenia Definitions and Outcomes Consortium (SDOC) supported by NIA/NIH, defined as "low muscle strength assessed by grip strength and slow gait speed" (ie, slowness)
• Per European Working Group on Sarcopenia in Older People 2 (EWGSOP2), defined as "low muscle mass, low muscle strength, and low physical performance"
Impaired proteostasis 20%
Frailty101,102 Clinical circumstance of increased vulnerability resulting from age-associated declines in reserve and function across multiple physiologic systems such that the ability to cope with everyday acute stress is compromised May be operationalized on the basis of physical attributes alone, (operationalized by Fried's criteria) or on the basis of an accumulation of deficits across multiple domains, calculated as a frailty index Mitochondrial dysfunction 50%
Cognitive impairment103 A limitation in the mental process of acquiring knowledge and understanding; can range from mild cognitive impairment to dementia Deregulated nutrient sensing 43% on the basis of a recent systematic review
a

Prevalence estimates vary based on numerous factors, including phenotype and severity of heart failure, diagnostic tool used, and study setting.

HF = heart failure; NIA = National Institute on Aging; NIH = National Institutes of Health.

GERIATRIC CONDITIONS AND CLINICAL DECISION MAKING.

Geriatric conditions complicate management in HF given their well-known associations with adverse outcomes, including impaired quality of life, hospitalization, and reduced life expectancy even after adjustment for chronologic age (Table 3). This provides strong rationale for the identification and integration of these conditions into clinical decision making and also clinical trials (in lieu of chronologic age).

TABLE 3.

Known Adverse Outcomes Associated With Geriatric Conditions Among Subgroups of Patients With HF

Malnutrition Cachexia Sarcopenia Frailty Cognitive Impairment

HFpEF ↑ Mortality
↑ Hospitalization
↑ Mortality
↑ Symptom burden
↓ QoL
↑ Mortality
↑ Hospitalization
↓ QoL
↑ Exercise capacity
↑ Mortality
↑ Hospitalization
↓ QoL
↑ Disability
↑ Mortality
↑ Hospitalization
↓ QoL
↑ Disability
HFrEF ↑ Mortality
↑ Hospitalization
↑ Mortality
↑ Symptom burden
↓ QoL
↑ Mortality
↑ Hospitalization
↑ Mortality
↑ Hospitalization
↓ QoL
↑ Disability
↑ Mortality
↑ Hospitalization
↓ QoL
↑ Disability
LVAD recipients ↑ Mortality
↑ ICU length of stay
↑ Sepsis
↑ Mortality ↑ Mortality
↑ GI bleeding
↑ Mortality
↑ Hospital LOS
↑ QoL
Transplant
recipients
↑ Mortality
↑ Need for MCS
↑ Sepsis
↑ Acute kidney injury
↑ Mortality ↑ Infection ↑ Mortality
↑ Hospital LOS
↑ Mortality

GI = gastrointestinal; ICU = intensive care unit; HFpEF = heart failure with preserved ejection fraction; HFrEF = heart failure with reduced ejection fraction; LOS = length of stay; LVAD = left ventricular assist device; MCS = mechanical circulatory support; QoL = quality of life.

Identifying geriatric conditions among older adults with advanced HF who are undergoing assessment for left ventricular assist device (LVAD) implantation and orthotopic cardiac transplantation is especially important. Given previous data showing that carefully selected older adults can benefit from these advanced therapies,106 systematic measurement of geriatric conditions that delineates biological age in individuals with advanced HF could improve these assessments.

Frailty and sarcopenia are particularly important to integrate into decision making for advanced therapies. Frailty is highly prevalent in patients considered for LVAD and orthotopic heart transplantation, irrespective of chronologic age, and it is associated with prolonged hospitalization and mortality in these individuals.107,108 Sarcopenia is also highly prevalent in HF.109 Preoperative measures of sarcopenia (through characteristics of the skeletal muscle on computed tomography scan) are associated with increased gastrointestinal bleeding and mortality among patients undergoing LVAD implantation,110 and they are an independent predictor of infection in patients undergoing orthotopic cardiac transplantation.111 These common clinical scenarios are illustrative of the importance of geriatric conditions in the clinical management of older adults with HF.

MANAGEMENT OF GERIATRIC CONDITIONS.

Managing geriatric conditions should be part of a comprehensive approach to caring for patients with HF.112 However, there is a paucity of data on the impact of interventions for HF on geriatric conditions. Table 4 outlines potential considerations for managing geriatric conditions when present, as well as interventions under study for directly treating these conditions. Given the close links among biological aging, geriatric conditions, and HF, interventions that improve outcomes in HF could also improve geriatric conditions and related measures. Table 5 summarizes the available data on the effect of HF-specific interventions on geriatric conditions and emerging patient-reported measures especially relevant to older adults. Unfortunately, there are limited data on the effect of HF-specific interventions on geriatric conditions and measures, because of both the systematic exclusion of older adults from many of the landmark studies in HF113and the lack of geriatric-specific metrics as relevant outcomes. By virtue of their higher baseline risk, the absolute benefits of HF-specific therapies are greatest among older adults, but this benefit can be offset by their attendant risks, which are often increased in the setting of geriatric conditions.112 To leverage emerging gerotherapeutic interventions in HF, future HF studies will need to include older adults with geriatric conditions and incorporate geriatric-specific metrics as end points.

TABLE 4.

Strategies and Potential Interventions for Managing Geriatric Conditions

Geriatric Condition Strategies for Enhanced Care Provision Interventions Requiring Additional Study

Malnutrition • Referral to dietitian/nutritionist
• Consideration of nutritional supplements
• Dietary recommendations (eg, increasing caloric intake and/or liberalizing dietary restrictions)
• Assessment of external factors that may be contributing to malnutrition (eg, financial means, taste, dental issues, social support)
• Dietary modifications
• Nutritional/caloric supplementation
• Combining nutritional interventions with exercise
• Home-delivered meals
• Micronutrient supplementation
Cachexia • Consideration of nutritional supplements
• Dietary recommendations (eg, increasing caloric intake and/or liberalizing dietary restrictions)
• Consideration for palliative care
• Treatment of underlying causes such as heart failure and other comorbid conditions
• Appetite stimulants
• Dietary modifications
• Nutritional/caloric supplementation
Sarcopenia • Referral to physical and/or occupational therapy; provision of exercise prescription
• Exercise and resistance training
• Reassessment of prognosis and risk-benefit ratio of management options
• Treatment of underlying causes such as heart failure (eg, effect of GDMT) and other comorbid conditions
• Nutritional/caloric supplementation
• Resistance exercise training
• Testosterone replacement
Frailty • Reassessment of prognosis and risk-benefit ratio of management options
• Emphasis on lifestyle recommendations such as exercise (home programs, cardiac rehabilitation programs, and strength-training) and nutrition
• Exercise
Cognitive impairment • Engagement of social support (caregivers, family), services
• Referral for formal assessment and/or discussion with other clinicians (geriatrics and/or memory center)
• Reassessment of health goals/priorities
• Reassessment of prognosis and risk-benefit ratio of management options (especially related to medications)
• Consideration of novel agents to treat early Alzheimer dementia if present
• Consideration for palliative care
• Treatment of underlying causes such as heart failure and other comorbid conditions
• Exercise

GDMT = guideline-directed medical therapy.

TABLE 5.

Effect of Heart Failure-Targeted Therapies on Select Geriatric Conditions and Measures

Effect on Select Geriatric Conditions
Effect on Select Geriatric Measures
Malnutrition/Cachexia Sarcopenia Frailty Cognitive Impairment Functional Capacitya Days Alive out of Hospital or Home Days ADL

Beta-adrenergic antagonists Attenuates and causes partial reversal No effect Unknown Unknown Worse in HFpEF (better with withdrawal) Improved in HFrEF Increased Unknown
ACEI/ARB Attenuates and causes partial reversal Unknown Unknown No effect Reduces rate of functional decline Increased Improved
ARNI Unknown Unknown Unknown No effect Improved Increased Improved compared with ACEI
Hydralazine/nitrates Unknown Unknown Unknown Unknown Improved Increased Unknown
MRAs Unknown Unknown Unknown Unknown No effect or improved Increased Unknown
SGLT2i Unknown Unknown Unknown Improved Improved Increased Unknown
GLP 1 RA Unknown Unknown Unknown Unknown Improved Unknown Unknown
TTR stabilizer Unknown Unknown Unknown Unknown Slows the decline Increased Unknown
Vericiguat Unknown Unknown No effect Unknown Unknown Increased Unknown
Cardiac rehabilitation Unknown Improved Improved Improved Improved Unknown Improved
 Aerobic training Unknown Improved Unknown Improved Improved No effect Unknown
 Resistance training Unknown Improved Unknown Unknown Improved Unknown Unknown
 Posthospitalization Unknown Improved Improved Unknown Improved Unknown Unknown
Cardiac resynchronization Unknown Unknown Unknown Improved Improved Increased Unknown
Cardiac contraction modulation Unknown Unknown Unknown Unknown Improved Unknown Unknown
Mitral valve clip Unknown Unknown Unknown Improved Improved Increased Unknown
Baroreflex simulation Unknown Unknown Unknown Unknown Improved Unknown Unknown
a

Functional improvements include changes in NYHA functional class, peak oxygen consumption, or 6-minute walk time.

ACEI = angiotensin-converting enzyme inhibitor; ADL = activities of daily living; ARB = angiotensin receptor blocker; ARNI = angiotensin blocker-neprilysin inhibitor; GLP 1 RA = glucagon-like peptide 1 receptoragonist; HFpEF = heart failure with preserved ejection fraction; HFrEF = heart failure with reduced ejection fraction; MRA = mineralocorticoid receptor antagonist; SGLT2i = sodium glucose cotransporter 2 inhibitor; TTR = transthyretin.

CONCLUSIONS AND FUTURE INVESTIGATIONS

The integration of geroscience into HF research not only is providing important new insights into the role of biological aging in HF, but also is beginning to change the therapeutic landscape for this chronic disease. For the field ultimately to leverage the potential of emerging gerotherapeutics in HF, additional work needs to be done to select candidate targets strategically on the basis of causal vs associative aging biology and then optimize implementation strategies, including appropriate patient populations in study design.

As highlighted throughout this review, some of the mechanisms underlying the hallmarks of aging are compensatory processes that are necessary for survival, but they become maladaptive when activated on a long-term basis or inhibited in the aging heart. In other words, not all biological aging is bad. This concept closely aligns with the neurohormonal model of HF pathophysiology, which has been a central tenet in HF therapeutic development for decades. In our effort to target biological aging therapeutically in HF, it is critical that we not only understand its role in physiological cardiovascular aging but also have a robust understanding of how direct manipulation (both gain and loss of function) of candidate targets affects cardiovascular aging and HF phenotypes. This will inevitably require rigorous investigations in preclinical models, not only across their natural life span, but also with targeted interventions at clinically relevant later stages of life in which many of these interventions would be implemented in older adults with HF.

Given the fundamental role of biological aging processes in cellular function and survival, along with the complex interactions among them, intervening on biological aging mechanisms likely will have broad effects. For both preclinical and clinical studies, comprehensive systemic phenotyping will be critical to assessing the safety and efficacy of emerging gerotherapeutics. In future HF trials, it will be important to include older adults more broadly and to integrate geriatric conditions into study design. Geriatric conditions such as frailty and cognitive impairment may be particularly relevant data to collect at baseline to understand better the effect of various pharmacologic and device-based interventions within subgroups of older adults with these conditions. Geriatric conditions could also potentially serve as important outcomes in clinical trials, given the importance of function and cognition on independence and overall well-being. Improved understanding about how various interventions interact with geriatric conditions could provide insights on underlying mechanisms that merit additional investigation.

HIGHLIGHTS.

  • Although chronologic age is on a fixed path, biological age may be modifiable.

  • Intervening on biological aging could improve on how we treat chronic diseases such as HF.

  • Impaired proteostasis, mitochondrial dysfunction, and deregulated nutrient sensing are targets in clinical gerotherapeutic studies.

  • Geriatric conditions are a window into aging processes, and they merit integration in clinical decision making and clinical trials in older adults with HF.

FUNDING SUPPORT AND AUTHOR DISCLOSURES

Dr Goyal has received grant support from National Institutes of Health (NIH) grants K76AG064428 and R21AG077092; and has received personal fees from Sensorum Health, Agepha Pharma, and Axon Therapies. Dr Maurer has received grant support from NIH grants R01HL139671 and R01AG081582-01; has received grants and personal fees from Alnylam, Pfizer, Eidos, Prothena, and Ionis; and has received personal fees from AstraZeneca, Akcea, Intellia, and Novo Nordisk. Dr Roh has received grant support from NIH grants K76AG064328 and R01HL170058, Hassenfeld Scholars Award, MGH Transformative Scholars Award, and the Fred and Ines Yeatts Fund for Innovative Research; and has received research support from Genentech, Amgen, and Keros Therapeutics.

ABBREVIATIONS AND ACRONYMS

AMPK

AMK-activated protein kinase

ATP

adenosine triphosphate

CPT

carnitine palmitoyltransferase

CVD

cardiovascular disease

FA

fatty acid

HFpEF

heart failure with preserved ejection fraction

HFrEF

heart failure with reduced ejection fraction

IGF

insulin-like growth factor

LVAD

left ventricular assist device

mTOR

mechanistic target of rapamycin

PGC-1

peroxisome proliferator-activated receptor gamma coactivator 1

PUFA

polyunsaturated fatty acid

ROS

reactive oxygen species

TTR

transthyretin

UPR

unfolded protein response

UPS

ubiquitin-proteasome system

REFERENCES

  • 1.Tsao CW, Aday AW, Almarzooq ZI, et al. Heart disease and stroke statistics-2023 update: a report from the American Heart Association. Circulation. 2023;147:e93–e621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pandey A, Kraus WE, Brubaker PH, Kitzman DW. Healthy aging and cardiovascular function: invasive hemodynamics during rest and exercise in 104 healthy volunteers. J Am Coll Cardiol HF. 2020;8:111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Roh J, Rhee J, Chaudhari V, Rosenzweig A. The role of exercise in cardiac aging: from physiology to molecular mechanisms. Circ Res. 2016;118:279–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–278. [DOI] [PubMed] [Google Scholar]
  • 5.Bozkurt B, Ahmad T, Alexander KM, et al. Heart failure epidemiology and outcomes statistics: a report of the Heart Failure Society of America. J Card Fail. 2023;29:1412–1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sierra F, Kohanski RA. The role of the national institute on aging in the development of the field of geroscience. Cold Spring Harb Perspect Med. 2023;13(10):a041211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Forman DE, Kuchel GA, Newman JC, et al. Impact of geroscience on therapeutic strategies for older adults with cardiovascular disease: JACC scientific statement. J Am Coll Cardiol. 2023;82:631–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ungvari Z, Tarantini S, Sorond F, Merkely B, Csiszar A. Mechanisms of vascular aging, a geroscience perspective: JACC focus seminar. J Am Coll Cardiol. 2020;75:931–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol. 2023;20(11):754–777. [DOI] [PubMed] [Google Scholar]
  • 11.Zhang W, Song M, Qu J, Liu GH. Epigenetic modifications in cardiovascular aging and diseases. Circ Res. 2018;123:773–786. [DOI] [PubMed] [Google Scholar]
  • 12.Fyhrquist F, Saijonmaa O, Strandberg T. The roles of senescence and telomere shortening in cardiovascular disease. Nat Rev Cardiol. 2013;10:274–283. [DOI] [PubMed] [Google Scholar]
  • 13.Hariharan N, Sussman MA. Cardiac aging - getting to the stem of the problem. J Mol Cell Cardiol. 2015;83:32–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Li H, Hastings MH, Rhee J, Trager LE, Roh JD, Rosenzweig A. Targeting age-related pathways in heart failure. Circ Res. 2020;126:533–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Owens WA, Walaszczyk A, Spyridopoulos I, Dookun E, Richardson GD. Senescence and senolytics in cardiovascular disease: promise and potential pitfalls. Mech Ageing Dev. 2021;198:111540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Liberale L, Badimon L, Montecucco F, Luscher TF, Libby P, Camici GG. Inflammation, aging, and cardiovascular disease: JACC review topic of the week. J Am Coll Cardiol. 2022;79:837–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tchkonia T, Palmer AK, Kirkland JL. New horizons: novel approaches to enhance healthspan through targeting cellular senescence and related aging mechanisms. J Clin Endocrinol Metab. 2021;106:e1481–e1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8:595–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ruberg FL, Berk JL. Transthyretin (TTR) cardiac amyloidosis. Circulation. 2012;126:1286–1300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vilchez D, Saez I, Dillin A. The role of protein clearance mechanisms in organismal ageing and age-related diseases. Nat Commun. 2014;5:5659. [DOI] [PubMed] [Google Scholar]
  • 21.Taneike M, Yamaguchi O, Nakai A, et al. Inhibition of autophagy in the heart induces age-related cardiomyopathy. Autophagy. 2010;6:600–606. [DOI] [PubMed] [Google Scholar]
  • 22.Wang X, Li J, Zheng H, Su H, Powell SR. Proteasome functional insufficiency in cardiac pathogenesis. Am J Physiol Heart Circ Physiol. 2011;301:H2207–H2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Eisenberg T, Abdellatif M, Schroeder S, et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat Med. 2016;22:1428–1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Blackwood EA, Azizi K, Thuerauf DJ, et al. Pharmacologic ATF6 activation confers global protection in widespread disease models by reprograming cellular proteostasis. Nat Commun. 2019;10:187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pattison JS, Sanbe A, Maloyan A, Osinska H, Klevitsky R, Robbins J. Cardiomyocyte expression of a polyglutamine preamyloid oligomer causes heart failure. Circulation. 2008;117:2743–2751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rajasekaran NS, Connell P, Christians ES, et al. Human alpha B-crystallin mutation causes oxidoreductive stress and protein aggregation cardiomyopathy in mice. Cell. 2007;130:427–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Buxbaum JN, Tagoe C, Gallo G, Walker JR, Kurian S, Salomon DR. Why are some amyloidoses systemic? Does hepatic “chaperoning at a distance” prevent cardiac deposition in a transgenic model of human senile systemic (transthyretin) amyloidosis? FASEB J. 2012;26:2283–2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Tannous P, Zhu H, Johnstone JL, et al. Autophagy is an adaptive response in desmin-related cardiomyopathy. Proc Natl Acad Sci U S A. 2008;105:9745–9750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hammarstrom P, Wiseman RL, Powers ET, Kelly JW. Prevention of transthyretin amyloid disease by changing protein misfolding energetics. Science. 2003;299:713–716. [DOI] [PubMed] [Google Scholar]
  • 30.Maurer MS, Schwartz JH, Gundapaneni B, et al. Tafamidis treatment for patients with transthyretin amyloid cardiomyopathy. N Engl J Med. 2018;379:1007–1016. [DOI] [PubMed] [Google Scholar]
  • 31.Maurer MS, Kale P, Fontana M, et al. Patisiran treatment in patients with transthyretin cardiac amyloidosis. N Engl J Med. 2023;389:1553–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gillmore JD, Gane E, Taubel J, et al. CRISPR-Cas9 In vivo gene editing for transthyretin amyloidosis. N Engl J Med. 2021;385:493–502. [DOI] [PubMed] [Google Scholar]
  • 33.Garcia-Pavia P, Aus dem Siepen F, Donal E, et al. Phase 1 trial of antibody NI006 for depletion of cardiac transthyretin amyloid. N Engl J Med. 2023;389:239–250. [DOI] [PubMed] [Google Scholar]
  • 34.Harman D Aging: a theory based on free radical and radiation chemistry. J Gerontol. 1956;11:298–300. [DOI] [PubMed] [Google Scholar]
  • 35.Butterfield DA, Halliwell B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat Rev Neurosci. 2019;20:148–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Short KR, Bigelow ML, Kahl J, et al. Decline in skeletal muscle mitochondrial function with aging in humans. Proc Natl Acad Sci U S A. 2005;102:5618–5623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhou B, Tian R. Mitochondrial dysfunction in pathophysiology of heart failure. J Clin Invest. 2018;128:3716–3726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kujoth GC, Hiona A, Pugh TD, et al. Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science. 2005;309:481–484. [DOI] [PubMed] [Google Scholar]
  • 39.Xie S, Xu SC, Deng W, Tang Q. Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications. Signal Transduct Target Ther. 2023;8:114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Trifunovic A, Wredenberg A, Falkenberg M, et al. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature. 2004;429:417–423. [DOI] [PubMed] [Google Scholar]
  • 41.Dai DF, Chen T, Wanagat J, et al. Age-dependent cardiomyopathy in mitochondrial mutator mice is attenuated by overexpression of catalase targeted to mitochondria. Aging Cell. 2010;9:536–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Vue Z, Neikirk K, Vang L, et al. Three-dimensional mitochondria reconstructions of murine cardiac muscle changes in size across aging. Am J Physiol Heart Circ Physiol. 2023;325:H965–H982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dai DF, Santana LF, Vermulst M, et al. Overexpression of catalase targeted to mitochondria attenuates murine cardiac aging. Circulation. 2009;119:2789–2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Arany Z, He H, Lin J, et al. Transcriptional coactivator PGC-1 alpha controls the energy state and contractile function of cardiac muscle. Cell Metab. 2005;1:259–271. [DOI] [PubMed] [Google Scholar]
  • 45.Arany Z, Novikov M, Chin S, Ma Y, Rosenzweig A, Spiegelman BM. Transverse aortic constriction leads to accelerated heart failure in mice lacking PPAR-gamma coactivator 1alpha. Proc Natl Acad Sci U S A. 2006;103:10086–10091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhu X, Shen W, Yao K, et al. Fine-Tuning of PGC1alpha expression regulates cardiac function and longevity. Circ Res. 2019;125:707–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, Kelly DP. Peroxisome proliferator-activated receptor gamma coactivator-1 promotes cardiac mitochondrial biogenesis. J Clin Invest. 2000;106:847–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.van der Pol A, van Gilst WH, Voors AA, van der Meer P. Treating oxidative stress in heart failure: past, present and future. Eur J Heart Fail. 2019;21:425–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sharma A, Fonarow GC, Butler J, Ezekowitz JA, Felker GM. Coenzyme Q10 and heart failure: a state-of-the-art review. Circ Heart Fail. 2016;9:e002639. [DOI] [PubMed] [Google Scholar]
  • 50.Skibska B, Goraca A. The protective effect of lipoic acid on selected cardiovascular diseases caused by age-related oxidative stress. Oxid Med Cell Longev. 2015;2015:313021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ochoa JJ, Quiles JL, Huertas JR, Mataix J. Coenzyme Q10 protects from aging-related oxidative stress and improves mitochondrial function in heart of rats fed a polyunsaturated fatty acid (PUFA)-rich diet. J Gerontol A Biol Sci Med Sci. 2005;60:970–975. [DOI] [PubMed] [Google Scholar]
  • 52.Mortensen SA, Rosenfeldt F, Kumar A, et al. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: results from Q-SYMBIO: a randomized double-blind trial. J Am Coll Cardiol HF. 2014;2:641–649. [DOI] [PubMed] [Google Scholar]
  • 53.Samuel TY, Hasin T, Gotsman I, et al. Coenzyme Q10 in the treatment of heart failure with preserved ejection fraction: a prospective, randomized, double-blind, placebo-controlled trial. Drugs R D. 2022;22:25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Monette JS, Gomez LA, Moreau RF, et al. ®-alpha-Lipoic acid treatment restores ceramide balance in aging rat cardiac mitochondria. Pharmacol Res. 2011;63:23–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Li CJ, Lv L, Li H, Yu DM. Cardiac fibrosis and dysfunction in experimental diabetic cardiomyopathy are ameliorated by alpha-lipoic acid. Cardiovasc Diabetol. 2012;11:73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hegazy SK, Tolba OA, Mostafa TM, Eid MA, El-Afify DR. Alpha-lipoic acid improves subclinical left ventricular dysfunction in asymptomatic patients with type 1 diabetes. Rev Diabet Stud. 2013;10:58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Tong D, Schiattarella GG, Jiang N, et al. NAD(+) Repletion Reverses Heart Failure With Preserved Ejection Fraction. Circ Res. 2021;128:1629–1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Abdellatif M, Trummer-Herbst V, Koser F, et al. Nicotinamide for the treatment of heart failure with preserved ejection fraction. Sci Transl Med. 2021;13:eabd7064. 10.1126/scitranslmed.abd7064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang DD, Airhart SE, Zhou B, et al. Safety and tolerability of nicotinamide riboside in heart failure with reduced ejection fraction. J Am Coll Cardiol Basic Trans Science. 2022;7:1183–1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Siegel MP, Kruse SE, Percival JM, et al. Mitochondrial-targeted peptide rapidly improves mitochondrial energetics and skeletal muscle performance in aged mice. Aging Cell. 2013;12:763–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Roshanravan B, Liu SZ, Ali AS, et al. In vivo mitochondrial ATP production is improved in older adult skeletal muscle after a single dose of elamipretide in a randomized trial. PLoS One. 2021;16:e0253849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Dai DF, Hsieh EJ, Chen T, et al. Global proteomics and pathway analysis of pressure-overload-induced heart failure and its attenuation by mitochondrial-targeted peptides. Circ Heart Fail. 2013;6:1067–1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sabbah HN, Gupta RC, Kohli S, Wang M, Hachem S, Zhang K. Chronic therapy with elamipretide (mtp-131), a novel mitochondria-targeting peptide, improves left ventricular and mitochondrial function in dogs with advanced heart failure. Circ Heart Fail. 2016;9:e002206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Butler J, Khan MS, Anker SD, et al. Effects of elamipretide on left ventricular function in patients with heart failure with reduced ejection fraction: the PROGRESS-HF phase 2 trial. J Card Fail. 2020;26:429–437. [DOI] [PubMed] [Google Scholar]
  • 65.Lee L, Campbell R, Scheuermann-Freestone M, et al. Metabolic modulation with perhexiline in chronic heart failure: a randomized, controlled trial of short-term use of a novel treatment. Circulation. 2005;112:3280–3288. [DOI] [PubMed] [Google Scholar]
  • 66.Beadle RM, Williams LK, Kuehl M, et al. Improvement in cardiac energetics by perhexiline in heart failure due to dilated cardiomyopathy. J Am Coll Cardiol HF. 2015;3:202–211. [DOI] [PubMed] [Google Scholar]
  • 67.Unger SA, Robinson MA, Horowitz JD. Perhexiline improves symptomatic status in elderly patients with severe aortic stenosis. Aust N Z J Med. 1997;27:24–28. [DOI] [PubMed] [Google Scholar]
  • 68.Stanley WC, Dabkowski ER, Ribeiro RF Jr, O’Connell KA. Dietary fat and heart failure: moving from lipotoxicity to lipoprotection. Circ Res. 2012;110:764–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Block RC, Liu L, Herrington DM, et al. Predicting risk for incident heart failure with omega-3 fatty acids: from MESA. J Am Coll Cardiol HF. 2019;7:651–661. [DOI] [PubMed] [Google Scholar]
  • 70.Chen J, Shearer GC, Chen Q, et al. Omega-3 fatty acids prevent pressure overload-induced cardiac fibrosis through activation of cyclic GMP/protein kinase G signaling in cardiac fibroblasts. Circulation. 2011;123:584–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tavazzi L, Maggioni AP, Marchioli R, et al. Effect of n-3 polyunsaturated fatty acids in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial. Lancet. 2008;372:1223–1230. [DOI] [PubMed] [Google Scholar]
  • 72.Einvik G, Klemsdal TO, Sandvik L, Hjerkinn EM. A randomized clinical trial on n-3 polyunsaturated fatty acids supplementation and all-cause mortality in elderly men at high cardiovascular risk. Eur J Cardiovasc Prev Rehabil. 2010;17:588–592. [DOI] [PubMed] [Google Scholar]
  • 73.Kim J, Wende AR, Sena S, et al. Insulin-like growth factor I receptor signaling is required for exercise-induced cardiac hypertrophy. Mol Endocrinol. 2008;22:2531–2543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Mann N, Rosenzweig A. Can exercise teach us how to treat heart disease? Circulation. 2012;126:2625–2635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chaanine AH, Hajjar RJ. AKT signalling in the failing heart. Eur J Heart Fail. 2011;13:825–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Vasan RS, Sullivan LM, D’Agostino RB, et al. Serum insulin-like growth factor I and risk for heart failure in elderly individuals without a previous myocardial infarction: the Framingham Heart Study. Ann Intern Med. 2003;139:642–648. [DOI] [PubMed] [Google Scholar]
  • 77.Abdellatif M, Trummer-Herbst V, Heberle AM, et al. Fine-tuning cardiac insulin-like growth factor 1 receptor signaling to promote health and longevity. Circulation. 2022;145:1853–1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sciarretta S, Forte M, Frati G, Sadoshima J. New insights into the role of mTOR signaling in the cardiovascular system. Circ Res. 2018;122:489–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Flynn JM, O’Leary MN, Zambataro CA, et al. Late-life rapamycin treatment reverses age-related heart dysfunction. Aging Cell. 2013;12:851–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dai DF, Karunadharma PP, Chiao YA, et al. Altered proteome turnover and remodeling by short-term caloric restriction or rapamycin rejuvenate the aging heart. Aging Cell. 2014;13:529–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Harrison DE, Strong R, Sharp ZD, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Weiss EP, Fontana L. Caloric restriction: powerful protection for the aging heart and vasculature. Am J Physiol Heart Circ Physiol. 2011;301:H1205–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Flanagan EW, Most J, Mey JT, Redman LM. Calorie restriction and aging in humans. Annu Rev Nutr. 2020;40:105–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kraus WE, Bhapkar M, Huffman KM, et al. 2 years of calorie restriction and cardiometabolic risk (CALERIE): exploratory outcomes of a multicentre, phase 2, randomised controlled trial. Lancet Diabetes Endocrinol. 2019;7:673–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Waziry R, Ryan CP, Corcoran DL, et al. Effect of long-term caloric restriction on DNA methylation measures of biological aging in healthy adults from the CALERIE trial. Nat Aging. 2023;3:248–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Riordan MM, Weiss EP, Meyer TE, et al. The effects of caloric restriction- and exercise-induced weight loss on left ventricular diastolic function. Am J Physiol Heart Circ Physiol. 2008;294:H1174–H1182. [DOI] [PubMed] [Google Scholar]
  • 87.Kitzman DW, Brubaker P, Morgan T, et al. Effect of caloric restriction or aerobic exercise training on peak oxygen consumption and quality of life in obese older patients with heart failure with preserved ejection fraction: a randomized clinical trial. JAMA. 2016;315:36–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mannick JB, Del Giudice G, Lattanzi M, et al. mTOR inhibition improves immune function in the elderly. Sci Transl Med. 2014;6:268ra179. [DOI] [PubMed] [Google Scholar]
  • 89.Singh M, Jensen MD, Lerman A, et al. Effect of low-dose rapamycin on senescence markers and physical functioning in older adults with coronary artery disease: results of a pilot study. J Frailty Aging. 2016;5:204–207. [DOI] [PubMed] [Google Scholar]
  • 90.Campbell JM, Bellman SM, Stephenson MD, Lisy K. Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: a systematic review and meta-analysis. Ageing Res Rev. 2017;40:31–44. [DOI] [PubMed] [Google Scholar]
  • 91.Kulkarni AS, Gubbi S, Barzilai N. Benefits of metformin in attenuating the hallmarks of aging. Cell Metab. 2020;32:15–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gundewar S, Calvert JW, Jha S, et al. Activation of AMP-activated protein kinase by metformin improves left ventricular function and survival in heart failure. Circ Res. 2009;104:403–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Li JZ, Li YR. Cardiovascular protection by metformin: latest advances in basic and clinical research. Cardiology. 2023;148:374–384. [DOI] [PubMed] [Google Scholar]
  • 94.Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin as a tool to target aging. Cell Metab. 2016;23:1060–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Schaum N, Lehallier B, Hahn O, et al. Ageing hallmarks exhibit organ-specific temporal signatures. Nature. 2020;583:596–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Tian YE, Cropley V, Maier AB, Lautenschlager NT, Breakspear M, Zalesky A. Heterogeneous aging across multiple organ systems and prediction of chronic disease and mortality. Nat Med. 2023;29:1221–1231. [DOI] [PubMed] [Google Scholar]
  • 97.Rebelo-Marques A, De Sousa Lages A, Andrade R, et al. aging hallmarks: the benefits of physical exercise. Front Endocrinol (Lausanne). 2018;9:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Roh JD, Houstis N, Yu A, et al. Exercise training reverses cardiac aging phenotypes associated with heart failure with preserved ejection fraction in male mice. Aging Cell. 2020;19:e13159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lerchenmuller C, Vujic A, Mittag S, et al. Restoration of cardiomyogenesis in aged mouse hearts by voluntary exercise. Circulation. 2022;146:412–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Werner C, Hanhoun M, Widmann T, et al. Effects of physical exercise on myocardial telomere-regulating proteins, survival pathways, and apoptosis. J Am Coll Cardiol. 2008;52:470–482. [DOI] [PubMed] [Google Scholar]
  • 101.Liu X, Xiao J, Zhu H, et al. miR-222 is necessary for exercise-induced cardiac growth and protects against pathological cardiac remodeling. Cell Metab. 2015;21:584–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Eschenhagen T, Bolli R, Braun T, et al. Cardiomyocyte regeneration: a consensus statement. Circulation. 2017;136:680–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.O’Connor CM, Whellan DJ, Lee KL, et al. Efficacy and safety of exercise training in patients with chronic heart failure: HF-ACTION randomized controlled trial. JAMA. 2009;301:1439–1450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kitzman DW, Whellan DJ, Duncan P, et al. Physical rehabilitation for older patients hospitalized for heart failure. N Engl J Med. 2021;385:203–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Inouye SK, Studenski S, Tinetti ME, Kuchel GA. Geriatric syndromes: clinical, research, and policy implications of a core geriatric concept. J Am Geriatr Soc. 2007;55:780–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Cooper LB, Lu D, Mentz RJ, et al. Cardiac transplantation for older patients: characteristics and outcomes in the septuagenarian population. J Heart Lung Transplant. 2016;35:362–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jha SR, Hannu MK, Chang S, et al. The prevalence and prognostic significance of frailty in patients with advanced heart failure referred for heart transplantation. Transplantation. 2016;100:429–436. [DOI] [PubMed] [Google Scholar]
  • 108.Macdonald PS, Gorrie N, Brennan X, et al. The impact of frailty on mortality after heart transplantation. J Heart Lung Transplant. 2021;40:87–94. [DOI] [PubMed] [Google Scholar]
  • 109.Springer J, Springer JI, Anker SD. Muscle wasting and sarcopenia in heart failure and beyond: update 2017. ESC Heart Fail. 2017;4:492–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Teigen LM, John R, Kuchnia AJ, et al. Preoperative pectoralis muscle quantity and attenuation by computed tomography are novel and powerful predictors of mortality after left ventricular assist device implantation. Circ Heart Fail. 2017;10(9):e004069. [DOI] [PubMed] [Google Scholar]
  • 111.Tsuji M, Kakuda N, Bujo C, et al. Sarcopenia and risk of infection in adult heart transplant recipients in Japan. ESC Heart Fail. 2022;9:1413–1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Gorodeski EZ, Goyal P, Hummel SL, et al. Domain management approach to heart failure in the geriatric patient: present and future. J Am Coll Cardiol. 2018;71:1921–1936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cherubini A, Oristrell J, Pla X, et al. The persistent exclusion of older patients from ongoing clinical trials regarding heart failure. Arch Intern Med. 2011;171:550–556. [DOI] [PubMed] [Google Scholar]

RESOURCES