Next Article in Journal
Lanthanides-Based Nanoparticles Conjugated with Rose Bengal for FRET-Mediated X-Ray-Induced PDT
Previous Article in Journal
Exploring the Protective Effects of Traditional Antidiabetic Medications and Novel Antihyperglycemic Agents in Diabetic Rodent Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity

by
Lauren A. Ling
1,2,
Asma Boukhalfa
1,
Andrew H. Kung
1,
Vicky K. Yang
3 and
Howard H. Chen
1,2,*
1
Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA
2
School of Medicine, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
3
Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Rd., North Grafton, MA 01536, USA
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(5), 671; https://doi.org/10.3390/ph18050671
Submission received: 31 March 2025 / Revised: 24 April 2025 / Accepted: 28 April 2025 / Published: 1 May 2025

Abstract

:
Autophagy, an evolutionarily conserved process, plays an important role in cellular homeostasis and human diseases. Cardiovascular dysfunction, which presents during cancer treatment or in cancer-free individuals years after treatment, is a growing clinical challenge. Millions of cancer survivors and patients face an unpredictable risk of developing cardiotoxicity. Cardiotoxicity due to cancer treatment, as well as cancer progression, has been linked to autophagy dysregulation. Modulating autophagy has been further proposed as a therapeutic treatment for both cancer and cardiovascular disorders. The safe and effective use of autophagy modulation as a cardioprotective strategy during cancer treatment especially requires careful consideration and experimentation to minimize the impact on cancer treatment. We focus here on recent advances in targeted autophagy modulation strategies that utilize interdisciplinary approaches in biomedical sciences and are potentially translatable to treat cardiotoxicity and improve cancer treatment outcomes. This review highlights non-small molecule autophagy modulators to enhance targeted therapy, nanomedicine for autophagy modulation and monitoring, and in vitro models and future experiments needed to bring novel autophagy discoveries from basic research to clinical translation.

1. Background

1.1. Cardio-Oncology: Cardiovascular Disease Due to Cancer Treatment

Cancer therapies, including chemotherapeutics, targeted drugs, and radiation, have all been indicated to have unwanted side effects affecting major organ systems. In the cardiovascular system, toxicity from cancer therapies can cause systolic dysfunction, hypertension, arrhythmia, myocarditis, heart failure, and in some cases, death [1]. For instance, in women 50 years or older who survive breast cancer, heart disease has now become the leading cause of death [2]. Cancer therapy-induced heart failure is particularly morbid, with an estimated 3.5 times greater risk of death compared to idiopathic cardiomyopathy [3]. The prognosis in these heart failure patients is even worse than those seen in ischemic cardiomyopathy patients [4]. In fact, chemotherapy-induced cardiotoxicity is the second leading cause of morbidity and mortality in cancer patients, with malignancy being the first [5]. The scope of the cardiotoxicity challenge has resulted in the emergence of a new field: cardio-oncology. Cardiotoxicity detected during cancer therapy often leads to a stoppage of treatment or a switch to a less effective option, negatively impacting outcomes. Better cardioprotective intervention strategies that do not interfere with therapy efficacy can thus improve the current treatment of cancer patients. However, the current knowledge gap limits the use of adjuvants that protect the cardiovascular system while preserving treatment efficacy, raising the urgent need for a deeper mechanistic understanding of cellular processes affected by cancer drugs and potential cardioprotective interventions.

1.2. Autophagy: A Well-Conserved Cellular Degradative and Recycling Process

A common pathway that differentially affects cardiovascular health and cancer is autophagy. Since the Nobel Prize was awarded in 2016 for the discovery of autophagy, many biological insights into the molecular and regulatory mechanisms of autophagy have been gained [6]. This review focuses on macroautophagy—hereafter referred to as autophagy—as other forms of autophagy such as microautophagy, chaperone-mediated autophagy, and substrate-specific autophagy are discussed in detail in other reviews [7,8,9]. Autophagy is a cellular homeostatic process conserved in virtually all mammalian cells to eliminate harmful stressors and recycle cellular constituents. Autophagy encompasses distinct stages: initiation, nucleation of the autophagosome, expansion and elongation of the autophagosome membrane, closure and fusion with the lysosome, and degradation of intravesicular products. Each stage has a subset of potential therapeutic targets for activating or inhibiting autophagy, achieved by genetic, pharmacological, or dietary interventions (Figure 1), providing potentially promising strategies for treating diverse human diseases [10,11]. During stress, dysfunctional organelles and proteins are sequestered into autophagosomes, trafficked to the lysosomes to form autophagolysosomes for degradation, and subsequently released into the cytoplasm for recycling. Autophagy is also intricately linked to cell death processes, including autophagy-mediated apoptosis (termed “autosis”), ferroptosis, and necroptosis, all of which are active areas of research to advance the treatment of cancer [12], as well as more recently in cardiovascular disease [13].
The autophagic process is dynamic [26,27]. The kinetics of autophagic flux, however, have not been characterized in disease states [28]. The biological impact of changes in autophagy on cell physiology and fate remains poorly characterized (Figure 2). The major limitations highlighted in this review are the following: (i) The lack of autophagy reporters that can track autophagy kinetics in real time [29]. Commonly deployed gold standards in autophagy detection, such as by fluorescent reporters of LC3, are not translatable to human clinical sample analysis. On the other hand, the biochemical analysis of autophagy biomarkers LC3, p62, and Beclin-1 can only accurately measure autophagic flux if repeated biopsy samples can be collected [30]. (ii) There are no suitable in vivo models that preserve the microenvironment, the heterogeneity of cell types in tissue, and cell–cell communication. (iii) Cardiotoxicity in cancer mouse models is still in its infancy. No translational model has been validated to investigate cardiotoxicity in large animal models, limiting the translation of the biological understanding gained at the bench to its impact in the human clinic. A multi-faceted interdisciplinary endeavor is thus needed to fully realize the potential of autophagy modulation to treat cancer and cardiovascular disease.

1.3. Autophagy Pathophysiology: Differential Impact of Autophagy Modulation on the Heart and Cancer

The biological impact of autophagy on cancer is relatively well studied. In the early stages of malignancy, defective autophagy promotes cancer initiation and progression as damaged organelles and proteins fail to be degraded, eliciting inflammation that modifies the tumor microenvironment and exacerbates cancer [31,32,33]. Paradoxically, in the later stages of malignancy, autophagy has been shown to promote tumorigenesis and metastasis by inducing neovascularization and other invasive properties including the secretion of pro-invasive cytokines, the detachment of metastatic cells, and the survival of cancer cells [34,35,36]. Excessive autophagy can either increase cancer cell death or increase unwanted chemoresistance [37,38]. Hence, autophagy may be a prognostic biomarker of cancer to better stage progression and tumorigenesis [39,40,41,42].
On the other hand, autophagy is an adaptive mechanism in the heart. The activation of cardioprotective autophagy and mitophagy in cardiomyocytes allows for the rapid degradation and recycling of intracellular stressors to maintain cell viability, and the effects of autophagy stimulation are well covered in previous reviews [43,44,45], demonstrating that the acute activation of autophagy exerts beneficial effects on cardiomyocytes in various forms of cardiomyopathy leading to heart failure. However, it remains controversial when and how autophagy modulation is safe as an adjuvant to minimize cardiovascular dysfunction in cancer patients.

1.4. Unrealized Therapeutic Potential of Autophagy Modulation to Simultaneously Treat Cancer and Cardiovascular Disease

Autophagy levels in cancer cells are highly dysregulated compared to those in non-cancerous cells [46]. Autophagy activation is likely to be beneficial in preventing the transformation of healthy cells into cancerous ones by clearing damaged organelles [47]. As tumors form and mature, autophagy may be activated in response to oxidative stress in the microenvironment to sustain tumor growth and metastasis [48]. Thus, it is unsurprising that autophagy inhibition by FDA-approved small molecules, such as chloroquine, aids in treating cancers at the bench and in clinical trials [49]. Many cancer treatments can also impact autophagy (Table 1). Paradoxically, treatments highly effective at mitigating cancer also activate autophagy [50]. Therefore, our understanding of the pathophysiological role of autophagy in cancer cells and the tumor response to cancer treatment remains incomplete.
Cancer drugs can induce the death of cancer cells via multiple autophagy-mediated apoptotic and non-apoptotic pathways, including autosis [72], ferroptosis [73], and necroptosis [74], which are emerging targets for novel cancer treatments. In fact, many drugs with efficacy against cancer have been shown to modulate autophagy, leading to the activation of cell death programming (Table 1). These same apoptotic and non-apoptotic pathways have also been linked to cardiovascular disease [75,76]. Accumulating evidence supports that treatments for HER2-positive breast cancer, including non-targeted chemotherapeutics, such as doxorubicin, and newer targeted cancer drugs, such as trastuzumab, can directly affect autophagy in cardiomyocytes and other cell types in the heart [77,78]. Autophagy dysregulation due to cancer drugs results in cardiomyopathy and heart failure, leading to systolic [55] or diastolic dysfunction [79], hypertension, and arrhythmia [80]. The mechanism underlying how autophagy dysregulation leads to cardiovascular dysfunction in the context of cancer and cancer treatment is, however, not well understood.
In the heart, the excessive up- or downregulation of autophagy can be detrimental to cells and impact cardiovascular health [81]. The heart is composed of a myriad of cell types, including contractile cardiomyocytes, endothelial cells, fibroblasts, immune cells, and others. The impact of autophagy levels on cell fate is cell type-dependent and disease-specific. In cardiomyocytes, autophagy is an adaptive mechanism that rapidly turns over intracellular stressors, such as damaged mitochondria or misfolded proteins, by recycling their constituents to enhance cell survival. In other words, autophagy plays an important role in maintaining cardiomyocyte homeostasis [82]. The activation of autophagy in cardiomyocytes has been reported to exert cardioprotective effects in a myriad of cardiovascular diseases, including cancer treatment-induced cardiotoxicity [83]. Cardiomyocyte autophagy activation through AMPK can reduce ROS levels, leading to the downregulation of ROS-induced NLRP3 inflammasome activation [84], a reported therapeutic target for doxorubicin-induced cardiotoxicity [85]. However, the activation of autophagy in coronary endothelial cells can induce endothelial progenitor cell apoptosis, inhibit cell proliferation and migration, and suppress reendothelialization [86], as seen in in vitro studies and in patients with coronary stents placed for atherosclerosis [87]. More recently, endothelial autophagy activation has been shown to attenuate the endothelial-to-mesenchymal transition [88], which in a rat model of chronic doxorubicin-induced cardiotoxicity activates the nuclear factor (NF)-κB pathway, leading to vascular injury and cardiac fibrosis [89]. Autophagy modulation can thus lead to further endothelial injury and unwanted inflammation. In cardiac fibroblasts, autophagy activation enhances the maturation of myofibroblasts and can regulate fibrosis to prevent adverse remodeling [90,91]. Yet, overstimulating fibroblast activation can lead to unwanted fibrosis. Taken together, a similar holistic approach [92] that leverages autophagy modulation can be promising to prevent or treat cardiovascular dysfunction during cancer treatment, but this needs to be carefully defined.
Much like in the case of the heart, tumors are composed of diverse cell types. Given the complex pathophysiological impact of autophagy on cancer and cardio-oncology, precision medicine approaches will likely be more effective, taking cancer stage, cancer type, and the cancer and cardiac responses to cancer treatment into consideration. Small molecules with robust autophagy-modulating effects have been studied extensively in preclinical cell and animal experiments and have been proposed to exert cardioprotective [93] or anti-cancer effects [94]. The signaling pathways or specific processes affected by autophagy modulators are often well established. For instance, rapamycin activates autophagy via the mTOR (mammalian target of rapamycin) pathway and has been shown to exert beneficial effects in cardiovascular diseases [95] as well as in cancer treatment [96]. Chloroquine, a well-established autophagy inhibitor that inhibits lysosomal fusion with autophagosomes, has been approved for clinical use in cancer treatments [49]. The impact of chloroquine on cardiotoxicity during cancer treatment, however, remains unknown.
A plethora of small molecule compounds capable of either activating or inhibiting autophagic processes are available and, in some cases, approved by the FDA. The established safety profile for human use certainly raises hope that the off-label use of drugs such as rapamycin, chloroquine, metformin, and statins as autophagy modulators can accelerate translation. However, concerns remain that autophagy modulation affects different cell types very differently and can often exert opposite effects from the desired outcome. The off-target effects of autophagy modulation thus require careful consideration when delivered systemically. This is currently a major limitation in advancing autophagy modulation to treat disease. Targeted modulation specific to the autophagy process or to specific cell types or organs is therefore sorely needed to leverage the full potential of autophagy as a therapy.

1.5. Non-Small Molecule Approaches to Achieve Targeted Autophagy Modulation

1.5.1. Nanoparticulates

Polymers of nanoparticulate constructs can be used to enhance the delivery of autophagy-modulating peptides and enhance their bioavailability to specific cells or organs. Since the description of a well-established autophagy-activating cell-penetrating Tat-Beclin 1 peptide composed of the human immunodeficiency virus 1’s transduction domain (Tat) fused to a conserved 18-amino acid sequence of Beclin 1 [97], further peptide modification strategies, particularly a diversity-oriented stapling approach, have been reported [98]. This approach produces an array of chemically circularized (“stapled”) Beclin 1-derived peptides to facilitate efficient cell penetration without the Tat sequence, and their diverse structures allow the diversity-oriented screening [99] of stapled peptides to isolate bioactive autophagy inducers. The most potent stapled peptide DD5-o at a micromolar concentration exerts autophagy-activating and aggregate-clearing activities in vitro in a cellular model of Huntington’s disease and induces autophagy in the muscles of mice in vivo [98]. Polylactic acid (PLA) particles surface-adsorbed with Tat-Beclin exhibit enhanced hepatic uptake in mice in vivo to induce autophagy and reduce lipid accumulation in an in vitro model of non-alcoholic fatty liver disease [100]. This confirms that not only is the Tat-Beclin peptide delivered intracellularly but it is also subsequently released and maintains its autophagy stimulating effects.
The cell-specific targeting of autophagy modulation is also feasible utilizing cell-type-specific targeting peptides that bind to the intended surface receptors. Cancer cell targeting is commonly achieved by RGD (Arginine–Glycine–Asparagine) peptides. For instance, a study demonstrated the synthesis of melanin-like polydopamine nanoparticles modified with dual RGD (which targets the alphavbeta3 integrin receptor on tumors) and Beclin 1-derived peptides (binds to intracellular Class III phosphatidylinositol 3-kinase) to induce autophagy in cancer cells [101]. The use of nanocarriers to modulate autophagy allows the pharmacodynamics and pharmacokinetics of autophagy modulation to be controlled, leading to better bioavailability. This is a potentially attractive option for translation.
While cancer-targeting peptides recognize overexpressed cancer cell receptors, cardiac targeting can be achieved by Cardiac Targeting Peptides. These peptides bind to potassium voltage-gated channel subfamily H member 5 (Kcnh5) to target murine and human cardiomyocytes [102]. The cardiomyocyte targeting of autophagy-modulating molecules or peptides, however, has not been reported. The specificity of these peptides to deliver autophagy-modulating molecules to cardiomyocytes compared to cancer cells warrants further investigation.
Interestingly, various nanoparticles have been shown to directly affect the autophagy process. Cerium oxide nanoparticles protect against doxorubicin-induced cardiotoxicity by increasing autophagic flux, myocardial cardiac enzymes, and myocardial contractility [103]. Graphene oxide (GO) is an ultrathin nanomaterial that has been shown to be capable of stimulating or inhibiting autophagy in cancer cells [104]. This suggests that while nanoparticles may be an attractive translational tool to enhance autophagy modulation when delivered systemically, the biological effects of nanoparticulates on specific cell types need to be carefully evaluated to achieve the desired autophagy-modulating effects.
Immune cells have been engineered directly to specifically modulate autophagy. For instance, in mice, macrophages have been engineered to induce LC3-dependent autophagy to enhance the efficacy of a Bacille Calmette–Guérin (BCG) vaccine, commonly used against tuberculosis [105]. Autophagy activation increases the antigen presentation of macrophages in the fight against tuberculosis. In another study with healthy human volunteers, autophagy was specifically induced in vaccine-induced antigen-specific CD8+ T cells [106]. The study provided evidence for the importance of autophagy in vaccine immunogenicity in older humans and uncovered two novel drug targets (translation factor eIF5A and transcription factor TFEB) that may increase vaccination efficiency in the elderly population. These studies raise intriguing possibilities to enhance immunotherapy by leveraging autophagy modulation and to further investigate its effects on cancer treatment as well as its associated cardiotoxicity.

1.5.2. Extracellular Vesicles

Naturally occurring circulating biomarkers, such as extracellular vesicles (EVs), are an emerging area of research in the diagnosis of cancer [107,108,109,110,111,112] and cardiotoxicity [113,114,115,116,117] and can also be engineered for therapeutic effects [118,119,120,121,122,123,124]. EVs are heterogeneous, nano-sized, double-membraned vesicles secreted endogenously by all cell types that can carry signaling molecules (e.g., lipids, nucleic acids, and proteins) and whole organelles [125]. EVs are important biomarkers of human disease that have also been explored as potential therapeutics. For instance, EVs from adipose-derived stromal cells were shown to contain miR-93-5p, which is responsible for inhibiting hypoxia-induced autophagy and inflammation and attenuates myocardial infarction in rats [126].
Additionally, EVs can modulate autophagy to protect tissues during periods of cellular stress [127]. Exosomes from induced pluripotent stem cell-derived cardiomyocytes promoted autophagosome production and autophagic flux, reduced apoptosis and fibrosis, and improved myocardial repair in mice [128]. Mesenchymal stem cell (MSC)-derived exosomes effectively protected hepatocytes against D-galactosamine and lipopolysaccharide (D-GalN/LPS)-induced damage via increasing levels of autophagic-related proteins (LC3B and Beclin-1) and the suppression of pro-apoptotic proteins [129]. Similar results were obtained in a rat model of spinal cord injury where the authors showed that neural stem cell-derived exosomes suppressed apoptosis and neuro-inflammation by inducing autophagy while reducing pro-inflammatory cytokine expression [130]. Exosomes from human MSCs also demonstrated the potential to attenuate ischemia–reperfusion injury by the inhibition of autophagy flux through the upregulation of mTORC1/p-4eBP1 [131]. However, the therapeutic potential of exosomes in preventing cardiac injury and the subsequent development of cardiomyopathy in cardio-oncology is not yet clear. Caution and further research are thus needed to harness the therapeutic potential of EVs to modulate autophagy in the target cell types.
More recently, autophagy has been implicated in the regulation of EVs [132,133,134]. Cargo sorting in EVs is achieved via autophagy machinery proteins [133]. Given that EV cargoes play an important role in their biological effect on recipient cells, it is important to understand the mechanism by which EV cargoes can specify the autophagy response, and how this response differs in cancer and cardiovascular cells under physiological and pathophysiological conditions.
Given that EVs are naturally occurring nanostructures, they have potential for translation. Further investigation into the impact of nanomaterials on the cardiovascular system and cancer will accelerate the translation of nanomedicine into the clinic as a novel class of biological molecules for targeted autophagy modulation.

1.5.3. mRNA

mRNA can also modulate autophagy, potentially opening the door for future targeted therapeutic strategies. Site-specific mRNA delivery methods are an emerging area of targeting strategies with increased payload delivery efficiency. These include the use of organ- or tissue-specific lipid nanoparticles (LNPs) following local injection, as well as organ- or cell-specific LNPs after intravenous injection [135]. mRNA was also functionalized on nanoparticles for delivery in a translational mouse model [136]. The mRNA nanoparticle-mediated restoration of PTEN in PTEN-null tumors has also shown that autophagy can be specifically induced to trigger cell death-associated immune activation. These mRNA-mediated autophagy modulation approaches can potentially support novel immunotherapies. However, potential off-target effects on other organs need to be carefully studied to overcome similar challenges faced by other mRNA-based therapies.

1.5.4. Other Strategies

More recently, cargo-specific autophagy has been explored to translate autophagy understanding into therapeutics. Autophagy-inhibiting peptides are useful in treating cancer [137]. Targeted peptides have been shown to maintain protein homeostasis and show specificity in modulating the proteosome pathway, which is distinct from macroautophagy [138]. The efficacy of these technologies is yet to be well tested in cancer and cardiovascular cells.
AceTAC degradation technology utilizes antigen–antibody interactions and autophagy to break down and remove intracellular stressors such as proteins [139]. Variations in this technology, such as AUTOTAC, utilize bifunctional small molecules to link p62 to specific targets, efficiently degrading selected proteins via autophagy [140]. More recently, the feasibility of the targeted degradation of organelles such as mitochondria, peroxisomes, and the endoplasmic reticulum in mammalian cells has been demonstrated [139]. Interestingly, cargo-specifying technology has also been explored to develop therapeutic strategies that “hijack” autophagy machinery to degrade specific cargoes. These areas of research may provide targeted autophagy therapeutics to tinker with the delicate balance in treating cardiotoxicity during cancer treatment.
In the era of artificial intelligence and big data, machine learning and system biological approaches have shed novel insights into autophagy modulation. Using a machine learning, cross-species workflow, eighteen small molecule autophagy modulators, including two novel mitophagy activators, were identified [141]. The authors further demonstrated that activating mitophagy was beneficial in mitigating memory loss and other disease pathologies in Alzheimer’s disease models of C. elegans and mice. A separate study using mathematical and computational modeling revealed that NRF2 (nuclear factor erythroid-related factor 2) interacts with the key autophagy signaling molecules AMPK and mTOR to fine-tune autophagy [142]. Autophagy induction by targeting AMPK, NRF2, and mTOR together can better adapt to dynamically changing environments in response to oxidative stress. Another study used mathematical modeling to predict and optimize drug dosing to achieve controlled autophagy modulation [143]. The authors demonstrated a generalizable strategy derived using computational methods to guide autophagy-based therapy selection with optimal drug scheduling.

1.6. Translational Models of Cardio-Oncology

To ensure the successful translation of the fundamental understanding of autophagy to improve the clinical management of human disease, cross-disciplinary collaboration among basic, translational, and clinical scientists is needed. The in vitro use of primary cardiomyocytes, whether of a human or large animal, however, remains a challenge as cardiomyocytes are terminally differentiated cells that are difficult to maintain in culture. Human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) have been well validated for studying cardiotoxicity [144,145,146]. However, autophagy is sensitive to culturing conditions and has not been carefully characterized in iPSCs. It remains to be determined whether autophagy and the response to autophagy activation or inhibition in iPSC-CMs recapitulate what is seen in primary cardiomyocytes.
Primary cardiac slices, on the other hand, are 100–400 μm viable tissue slices generated from a living adult ventricular myocardium using a high-precision vibratome system [147,148]. The slices represent a suitable in vitro system that provides several major advantages over isolated cardiomyocytes: (i) the cardiovascular architecture and cell types are preserved; (ii) cardiac slices have been shown to survive in culture for one week; and (iii) the slices can be generated from all animal models as well as human tissue [147,148,149,150,151]. In addition, basal autophagy and the response to autophagy modulations in cardiac slices were recently validated in canine slices [147]. Cardiac slices are also amenable to co-culturing with cancer cells, 3D cancer organoids, and immune cells, creating a more realistic cancer and treatment setting more closely mimicking an in vivo scenario. This is critical in the context of cardio-oncology to understand the cardiac autophagy response to the presence of a tumor and the associated pro-inflammatory environment with or without the addition of chemotherapy and autophagy modulation. The cardiac response to chemotherapy with or without autophagy modulation is likely influenced by intercellular and inter-tissue or organ communications seen in the in vivo environment. Cardiac slices generated from healthy or pathological hearts isolated from large animals such as the canine provide a uniquely powerful in vitro model in drug development. The slice system thus presents an ideal preclinical evaluation tool for the high-throughput screening of drugs that may yield lead compounds to be further tested in canine models.
An appropriate translational animal model for in vivo cardio-oncology research remains lacking. Preclinical transgenic, humanized, and implanted tumor mouse models that are commonly used often do not faithfully recapitulate the complex and heterogeneous cancer phenotype in human patients [152,153]. This further limits the study of cancer treatment-induced cardiotoxicity in murine models [154,155]. Unlike mice, which do not spontaneously develop cancer, canines are particularly relevant in the study of human cancer and the cancer response to drugs as they develop spontaneous cancer much like humans and receive the same or similar cancer therapies and cancer drugs as humans. For example, it is well established that dogs receiving anthracycline develop cardiotoxicity manifesting as systolic dysfunction, heart failure, and arrhythmias, similar to human cancer patients receiving anthracycline therapy [156]. These canine patients also develop systemic hypertension in response to tyrosine kinase inhibitor treatments, similar to responses seen in human cancer patients [157]. Clinical trials in pet dogs undergoing cancer treatment thus present a uniquely suitable translational model to study responses associated with autophagy modulation, cancer treatment, and cardiotoxicity.

1.7. Autophagy Monitoring Technology

There is currently a lack of suitable biomarkers to follow the onset and progress of cardiotoxicity during cancer therapy. Given the pathophysiological significance of autophagy in both cancers and the heart, validating autophagy as a potentially viable prognostic biomarker in the context of cardio-oncology warrants further investigation. In healthy cells, basal autophagy involves the intricate intracellular trafficking of autophagic vesicles, which is highly dynamic and tightly regulated [33,158]. During disease states, the dysregulation of autophagy alters the kinetics of autophagic activity (referred to as autophagic flux). To accurately assess autophagic flux, autophagy must be studied longitudinally or measured at multiple time points. Fluorescent reporters of autophagy proteins such as the dual fluorescent LC3 (Light chain 3, a marker of autophagic structures destined for degradation and recycling) plasmid have allowed autophagic flux to be measured in real time in vitro [159,160,161]. Transgenic mice ubiquitously expressing genetic fluorescent reporters of autophagy are also well established for studying autophagy in vivo [149,162,163]. Engineering florescent autophagy reporter mice with cancer allows autophagy in cancer and cardiotoxicity to be studied in the in vivo environment.
The real-time monitoring of autophagy levels non-invasively in vivo was recently demonstrated [164]. Autophagy-detecting nanoparticles (ADNs), rationally designed to translocate into cells efficiently, co-traffic with early autophagosomes to lysosomes, allowing autophagy to be imaged with high sensitivity and specificity, and are quantitative of autophagic flux [164]. Probe-based autophagy imaging using injectable nanoprobes such as ADNs emits activatable near-infrared fluorescence, allowing autophagy to be detected and quantified in cells in vitro and in small animal models non-invasively in vivo [164]. An ADN injected intravenously reaches all major organs, allowing longitudinal and systemic autophagy monitoring to optimize autophagy modulation in the preclinical context. In other words, this allows a holistic approach to guide autophagy modulation that maintains homeostasis in the heart while maintaining cancer treatment efficacy.
Additionally, the ADN is biocompatible, synthesized based on FDA-approved ferumoxytol, a drug used to treat amenia. Ferumoxytol in humans has a blood half-life of 14 to 21 h and is eliminated via the hepatic and splenic routes [165]. The ADN produces activatable magnetic resonance signals, detectable by non-invasive medical imaging modalities such as magnetic resonance imaging (MRI). The ADN-enhanced imaging of autophagy needs further validation in translational animal models. Given the biomedical significance of autophagy as a viable biomarker, further developing autophagy imaging translational to the clinic is a highly novel and worthwhile endeavor.

2. Conclusions and Future Outlooks

Autophagy modulation as a therapeutic strategy has tremendous potential, but obstacles remain for clinical adaptation. The field of cardio-oncology presents particularly unique challenges as well as opportunities to leverage the power of autophagy biology and innovations in autophagy modulation to revolutionize the treatment of cancer and cancer therapy-associated cardiotoxicity. A deeper biological understanding is needed to untangle the differential impact of autophagy activation or inhibition on cardiovascular health while preserving or, in ideal cases, enhancing cancer therapy efficacy.
This review highlights non-small molecule approaches to modulate autophagy, including engineered peptides that target specific autophagy-regulating proteins or nanoformulations capable of increasing bioavailability to the target cell type or organ. Nanomedicine has been an active area of research for cancer therapy [166,167,168] and may be a novel approach to mitigating cardiotoxicity by targeting autophagy modulation [169,170,171]. Nanotechnology further allows the probe-based monitoring of autophagy levels non-invasively in vivo [164]. Moreover, novel translation model systems are now available for new cancer drug testing to bridge preclinical investigations to the clinical setting. These highlights are summarized in Figure 3.
Future research directions to overcome the hurdles of translating autophagy modulation into clinical practice are sevenfold: (a) Biomarker discovery, likely from non-coding RNAs [172] or multi-omics studies [173], can identify specific biomarkers that could predict the effectiveness of autophagy modulation to guide cancer treatment while preventing cardiotoxicity. (b) The further development of molecular imaging of autophagy non-invasively in vivo can open novel portals into dynamic autophagy changes in the heart and tumors during cancer treatment. (c) The gain of a deeper mechanistic and pharmacological understanding in biological systems or the optimization of drug delivery methodologies can minimize the off-target effects of both small-molecule and non-small-molecule autophagy modulators. (d) Drug screening platforms that are amenable for translation must be enhanced. (e) Finally, multidisciplinary approaches through collaborations among basic scientists, translational researchers, and clinical cardiologists and oncologists must be integrated. Further autophagy research promises to treat cardiovascular disease and cancer with precision and provide real-time monitoring of treatment responses to improve outcomes. The research endeavor will not only impact the field of cardio-oncology, but it will also improve the treatment of a plethora of human diseases where autophagy dysregulation plays a role.

Author Contributions

All authors (L.A.L., A.B., A.H.K., V.K.Y. and H.H.C.) contributed to the writing, reviewing, and editing of the manuscript. L.A.L. and A.H.K. contributed to the generation of the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Institute of Health: K99/R00-HL121152 (HHC), R01-HL166810 (HHC), K01OD028205 (VKY), and the American Heart Association: CDA-932792 (HHC), 24POST1243937 (AB).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

MTORmammalian target of rapamycin
AMPKAMP-activated protein kinase
FDAU.S. Food and Drug Administration
V-ATPasevacuolar-type H+-ATPase
TFEBtranscription factor EB
LC3light chain 3
TATtransactivator of transcription
PLApolylactic acid particles
RGDArginine–Glycine–Asparagine peptide
Kcnh5potassium voltage-gated channel subfamily H member 5
GOGraphene oxide
BCGBacille Calmette–Guérin
EIF5Aeukaryotic translation initiation factor 5A
EVextracellular vesicle
D-GalND-galactosamine
LPSlipopolysaccharide
MSCmesenchymal stem cell
p-4eBP1phosphorylated translation initiation factor 4E-binding protein 1
LNPlipid nanoparticle
PTENphosphatase and tensin homolog
AceTACautophagy receptor-inspired targeting chimera
AUTOTACautophagy-targeting chimera
NRF2nuclear factor erythroid-related factor 2
iPSC-CMshuman-induced pluripotent stem cell-derived cardiomyocytes
ADNautophagy-detecting nanoparticle
MRImagnetic resonance imaging

References

  1. Herrmann, J. Adverse cardiac effects of cancer therapies: Cardiotoxicity and arrhythmia. Nat. Rev. Cardiol. 2020, 17, 474–502. [Google Scholar] [CrossRef] [PubMed]
  2. Garcia, M.; Mulvagh, S.L.; Merz, C.N.; Buring, J.E.; Manson, J.E. Cardiovascular Disease in Women: Clinical Perspectives. Circ. Res. 2016, 118, 1273–1293. [Google Scholar] [CrossRef] [PubMed]
  3. Bloom, M.W.; Hamo, C.E.; Cardinale, D.; Ky, B.; Nohria, A.; Baer, L.; Skopicki, H.; Lenihan, D.J.; Gheorghiade, M.; Lyon, A.R.; et al. Cancer Therapy-Related Cardiac Dysfunction and Heart Failure: Part 1, Definitions, Pathophysiology, Risk Factors, and Imaging. Circ. Heart Fail. 2016, 9, e002661. [Google Scholar] [CrossRef]
  4. Felker, G.M.; Thompson, R.E.; Hare, J.M.; Hruban, R.H.; Clemetson, D.E.; Howard, D.L.; Hruban, R.H.; Clemetson, D.E.; Howard, D.L.; Baughman, K.L.; et al. Underlying causes and long-term survival in patients with initially unexplained cardiomyopathy. N. Engl. J. Med. 2000, 342, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
  5. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef]
  6. Levine, B.; Kroemer, G. Biological Functions of Autophagy Genes: A Disease Perspective. Cell 2019, 176, 11–42. [Google Scholar] [CrossRef]
  7. Kaushik, S.; Cuervo, A.M. The coming of age of chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 365–381. [Google Scholar] [CrossRef]
  8. Parzych, K.R.; Klionsky, D.J. An overview of autophagy: Morphology, mechanism, and regulation. Antioxid. Redox Signal. 2014, 20, 460–473. [Google Scholar] [CrossRef]
  9. Wang, L.; Klionsky, D.J.; Shen, H.M. The emerging mechanisms and functions of microautophagy. Nat. Rev. Mol. Cell Biol. 2023, 24, 186–203. [Google Scholar] [CrossRef]
  10. Rubinsztein, D.C.; Codogno, P.; Levine, B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat. Rev. Drug Discov. 2012, 11, 709–730. [Google Scholar] [CrossRef]
  11. Vakifahmetoglu-Norberg, H.; Xia, H.G.; Yuan, J. Pharmacologic agents targeting autophagy. J. Clin. Investig. 2015, 125, 5–13. [Google Scholar] [CrossRef] [PubMed]
  12. Mulcahy Levy, J.M.; Thorburn, A. Autophagy in cancer: Moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ. 2020, 27, 843–857. [Google Scholar] [CrossRef]
  13. Nemchenko, A.; Chiong, M.; Turer, A.; Lavandero, S.; Hill, J.A. Autophagy as a therapeutic target in cardiovascular disease. J. Mol. Cell. Cardiol. 2011, 51, 584–593. [Google Scholar] [CrossRef] [PubMed]
  14. Sarkar, S.; Ravikumar, B.; Floto, R.A.; Rubinsztein, D.C. Rapamycin and mTOR-independent autophagy inducers ameliorate toxicity of polyglutamine-expanded huntingtin and related proteinopathies. Cell Death Differ. 2009, 16, 46–56. [Google Scholar] [CrossRef]
  15. Schreiber, K.H.; Ortiz, D.; Academia, E.C.; Anies, A.C.; Liao, C.Y.; Kennedy, B.K. Rapamycin-mediated mTORC2 inhibition is determined by the relative expression of FK506-binding proteins. Aging Cell 2015, 14, 265–273. [Google Scholar] [CrossRef] [PubMed]
  16. Cai, Y.; Xia, Q.; Su, Q.; Luo, R.; Sun, Y.; Shi, Y.; Jiang, W. mTOR inhibitor RAD001 (everolimus) induces apoptotic, not autophagic cell death, in human nasopharyngeal carcinoma cells. Int. J. Mol. Med. 2013, 31, 904–912. [Google Scholar] [CrossRef]
  17. Zhao, J.; Zhai, B.; Gygi, S.P.; Goldberg, A.L. mTOR inhibition activates overall protein degradation by the ubiquitin proteasome system as well as by autophagy. Proc. Natl. Acad. Sci. USA 2015, 112, 15790–15797. [Google Scholar] [CrossRef]
  18. Shang, L.; Chen, S.; Du, F.; Li, S.; Zhao, L.; Wang, X. Nutrient starvation elicits an acute autophagic response mediated by Ulk1 dephosphorylation and its subsequent dissociation from AMPK. Proc. Natl. Acad. Sci. USA 2011, 108, 4788–4793. [Google Scholar] [CrossRef]
  19. Tomic, T.; Botton, T.; Cerezo, M.; Robert, G.; Luciano, F.; Puissant, A.; Gounon, P.; Allegra, M.; Bertolotto, C.; Bereder, J.M.; et al. Metformin inhibits melanoma development through autophagy and apoptosis mechanisms. Cell Death Dis. 2011, 2, e199. [Google Scholar] [CrossRef]
  20. Rusmini, P.; Cortese, K.; Crippa, V.; Cristofani, R.; Cicardi, M.E.; Ferrari, V.; Vezzoli, G.; Tedesco, B.; Meroni, M.; Messi, E.; et al. Trehalose induces autophagy via lysosomal-mediated TFEB activation in models of motoneuron degeneration. Autophagy 2019, 15, 631–651. [Google Scholar] [CrossRef]
  21. Zhu, L.; Wang, Y.; Lv, W.; Wu, X.; Sheng, H.; He, C.; Hu, J. Schizandrin A can inhibit non-small cell lung cancer cell proliferation by inducing cell cycle arrest, apoptosis and autophagy. Int. J. Mol. Med. 2021, 48, 214. [Google Scholar] [CrossRef]
  22. Wang, G.; Wang, T.; Zhang, Y.; Li, F.; Yu, B.; Kou, J. Schizandrin Protects against OGD/R-Induced Neuronal Injury by Suppressing Autophagy: Involvement of the AMPK/mTOR Pathway. Molecules 2019, 24, 3624. [Google Scholar] [CrossRef] [PubMed]
  23. Mauthe, M.; Orhon, I.; Rocchi, C.; Zhou, X.; Luhr, M.; Hijlkema, K.J.; Coppes, R.P.; Engedal, N.; Mari, M.; Reggiori, F. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy 2018, 14, 1435–1455. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, R.; Wang, J.; Hassan, A.; Lee, C.H.; Xie, X.S.; Li, X. Molecular basis of V-ATPase inhibition by bafilomycin A1. Nat. Commun. 2021, 12, 1782. [Google Scholar] [CrossRef]
  25. Ahsan, A.; Zheng, Y.; Ma, S.; Liu, M.; Cao, M.; Li, Y.; Zheng, W.; Zhou, X.; Xin, M.; Hu, W.W.; et al. Tomatidine protects against ischemic neuronal injury by improving lysosomal function. Eur. J. Pharmacol. 2020, 882, 173280. [Google Scholar] [CrossRef] [PubMed]
  26. Kimmelman, A.C. The dynamic nature of autophagy in cancer. Genes Dev. 2011, 25, 1999–2010. [Google Scholar] [CrossRef]
  27. Shibutani, S.T.; Yoshimori, T. A current perspective of autophagosome biogenesis. Cell Res. 2014, 24, 58–68. [Google Scholar] [CrossRef]
  28. Loos, B.; du Toit, A.; Hofmeyr, J.H. Defining and measuring autophagosome flux-concept and reality. Autophagy 2014, 10, 2087–2096. [Google Scholar] [CrossRef]
  29. Bishnu, A.; Phadte, P.; Dhadve, A.; Sakpal, A.; Rekhi, B.; Ray, P. Molecular imaging of the kinetics of hyperactivated ERK1/2-mediated autophagy during acquirement of chemoresistance. Cell Death Dis. 2021, 12, 161. [Google Scholar] [CrossRef]
  30. Mizushima, N.; Murphy, L.O. Autophagy Assays for Biological Discovery and Therapeutic Development. Trends Biochem. Sci. 2020, 45, 1080–1093. [Google Scholar] [CrossRef]
  31. Balkwill, F.R.; Mantovani, A. Cancer-related inflammation: Common themes and therapeutic opportunities. Semin. Cancer Biol. 2012, 22, 33–40. [Google Scholar] [CrossRef] [PubMed]
  32. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
  33. Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef] [PubMed]
  34. Debnath, J.; Mills, K.R.; Collins, N.L.; Reginato, M.J.; Muthuswamy, S.K.; Brugge, J.S. The role of apoptosis in creating and maintaining luminal space within normal and oncogene-expressing mammary acini. Cell 2002, 111, 29–40. [Google Scholar] [CrossRef]
  35. Fung, C.; Lock, R.; Gao, S.; Salas, E.; Debnath, J. Induction of autophagy during extracellular matrix detachment promotes cell survival. Mol. Biol. Cell 2008, 19, 797–806. [Google Scholar] [CrossRef]
  36. Patergnani, S.; Missiroli, S.; Morciano, G.; Perrone, M.; Mantovani, C.M.; Anania, G.; Fiorica, F.; Pinton, P.; Giorgi, C. Understanding the Role of Autophagy in Cancer Formation and Progression Is a Real Opportunity to Treat and Cure Human Cancers. Cancers 2021, 13, 5622. [Google Scholar] [CrossRef]
  37. Fulda, S.; Kogel, D. Cell death by autophagy: Emerging molecular mechanisms and implications for cancer therapy. Oncogene 2015, 34, 5105–5113. [Google Scholar] [CrossRef]
  38. Niklaus, N.J.; Tokarchuk, I.; Zbinden, M.; Schlafli, A.M.; Maycotte, P.; Tschan, M.P. The Multifaceted Functions of Autophagy in Breast Cancer Development and Treatment. Cells 2021, 10, 1447. [Google Scholar] [CrossRef]
  39. He, Q.; Li, Z.; Yin, J.; Li, Y.; Yin, Y.; Lei, X.; Zhu, W. Prognostic Significance of Autophagy-Relevant Gene Markers in Colorectal Cancer. Front. Oncol. 2021, 11, 566539. [Google Scholar] [CrossRef]
  40. Jin, W.; Wu, L.; Hu, L.; Fu, Y.; Fan, Z.; Mou, Y.; Ma, K. Multi-omics approaches identify novel prognostic biomarkers of autophagy in uveal melanoma. J. Cancer Res. Clin. Oncol. 2023, 149, 16691–16703. [Google Scholar] [CrossRef]
  41. Peng, S.; Ma, S.; Yang, F.; Xu, C.; Li, H.; Lu, S.; Zhang, J.; Jiao, J.; Han, D.; Shi, C.; et al. Prognostic value and underlying mechanism of autophagy-related genes in bladder cancer. Sci. Rep. 2022, 12, 2219. [Google Scholar] [CrossRef] [PubMed]
  42. Vitto, V.A.M.; Bianchin, S.; Zolondick, A.A.; Pellielo, G.; Rimessi, A.; Chianese, D.; Yang, H.; Carbone, M.; Pinton, P.; Giorgi, C.; et al. Molecular Mechanisms of Autophagy in Cancer Development, Progression, and Therapy. Biomedicines 2022, 10, 1596. [Google Scholar] [CrossRef] [PubMed]
  43. Bravo-San Pedro, J.M.; Kroemer, G.; Galluzzi, L. Autophagy and Mitophagy in Cardiovascular Disease. Circ. Res. 2017, 120, 1812–1824. [Google Scholar] [CrossRef]
  44. Gatica, D.; Chiong, M.; Lavandero, S.; Klionsky, D.J. The role of autophagy in cardiovascular pathology. Cardiovasc. Res. 2022, 118, 934–950. [Google Scholar] [CrossRef]
  45. Titus, A.S.; Sung, E.A.; Zablocki, D.; Sadoshima, J. Mitophagy for cardioprotection. Basic Res. Cardiol. 2023, 118, 42. [Google Scholar] [CrossRef] [PubMed]
  46. Verma, A.K.; Bharti, P.S.; Rafat, S.; Bhatt, D.; Goyal, Y.; Pandey, K.K.; Ranjan, S.; Almatroodi, S.A.; Alsahli, M.A.; Rahmani, A.H.; et al. Autophagy Paradox of Cancer: Role, Regulation, and Duality. Oxidative Med. Cell. Longev. 2021, 2021, 8832541. [Google Scholar] [CrossRef]
  47. White, E.; Mehnert, J.M.; Chan, C.S. Autophagy, Metabolism, and Cancer. Clin. Cancer Res. 2015, 21, 5037–5046. [Google Scholar] [CrossRef]
  48. Mowers, E.E.; Sharifi, M.N.; Macleod, K.F. Autophagy in cancer metastasis. Oncogene 2017, 36, 1619–1630. [Google Scholar] [CrossRef]
  49. Mohsen, S.; Sobash, P.T.; Algwaiz, G.F.; Nasef, N.; Al-Zeidaneen, S.A.; Karim, N.A. Autophagy Agents in Clinical Trials for Cancer Therapy: A Brief Review. Curr. Oncol. 2022, 29, 1695–1708. [Google Scholar] [CrossRef]
  50. Thorburn, A.; Thamm, D.H.; Gustafson, D.L. Autophagy and cancer therapy. Mol. Pharmacol. 2014, 85, 830–838. [Google Scholar] [CrossRef]
  51. Li, D.L.; Wang, Z.V.; Ding, G.; Tan, W.; Luo, X.; Criollo, A.; Xie, M.; Jiang, N.; May, H.; Kyrychenko, V.; et al. Doxorubicin Blocks Cardiomyocyte Autophagic Flux by Inhibiting Lysosome Acidification. Circulation 2016, 133, 1668–1687. [Google Scholar] [CrossRef] [PubMed]
  52. Xu, J.; Gewirtz, D.A. Is Autophagy Always a Barrier to Cisplatin Therapy? Biomolecules 2022, 12, 463. [Google Scholar] [CrossRef]
  53. Xiao, B.; Hong, L.; Cai, X.; Mei, S.; Zhang, P.; Shao, L. The true colors of autophagy in doxorubicin-induced cardiotoxicity. Oncol. Lett. 2019, 18, 2165–2172. [Google Scholar] [CrossRef] [PubMed]
  54. Lin, J.F.; Lin, Y.C.; Tsai, T.F.; Chen, H.E.; Chou, K.Y.; Hwang, T.I. Cisplatin induces protective autophagy through activation of BECN1 in human bladder cancer cells. Drug Des. Devel. Ther. 2017, 11, 1517–1533. [Google Scholar] [CrossRef]
  55. Schirone, L.; D’Ambrosio, L.; Forte, M.; Genovese, R.; Schiavon, S.; Spinosa, G.; Iacovone, G.; Valenti, V.; Frati, G.; Sciarretta, S. Mitochondria and Doxorubicin-Induced Cardiomyopathy: A Complex Interplay. Cells 2022, 11, 2000. [Google Scholar] [CrossRef] [PubMed]
  56. Alvarez-Meythaler, J.G.; Garcia-Mayea, Y.; Mir, C.; Kondoh, H.; ME, L.L. Autophagy Takes Center Stage as a Possible Cancer Hallmark. Front. Oncol. 2020, 10, 586069. [Google Scholar] [CrossRef]
  57. Kadry, M.O.; Ammar, N.M.; Hassan, H.A.; Abdel Megeed, R.M. Insights on attenuating autophagy cellular and molecular pathways versus methotrexate-induced toxicity via liposomal turmeric therapy. J. Genet. Eng. Biotechnol. 2022, 20, 147. [Google Scholar] [CrossRef]
  58. Liu, Y.; Levine, B. Autosis and autophagic cell death: The dark side of autophagy. Cell Death Differ. 2015, 22, 367–376. [Google Scholar] [CrossRef]
  59. Xia, P.; Chen, J.; Liu, Y.; Fletcher, M.; Jensen, B.C.; Cheng, Z. Doxorubicin induces cardiomyocyte apoptosis and atrophy through cyclin-dependent kinase 2-mediated activation of forkhead box. OJ Biol. Chem. 2020, 295, 4265–4276. [Google Scholar] [CrossRef]
  60. Hu, Y.; Sun, B.; Zhao, B.; Mei, D.; Gu, Q.; Tian, Z. Cisplatin-induced cardiotoxicity with midrange ejection fraction: A case report and review of the literature. Medicine 2018, 97, e13807. [Google Scholar] [CrossRef]
  61. Baoqi, Y.; Dan, M.; Xingxing, Z.; Xueqing, Z.; Yajing, W.; Ke, X.; Liyun, Z. Effect of Anti-Rheumatic Drugs on Cardiovascular Disease Events in Rheumatoid Arthritis. Front. Cardiovasc. Med. 2021, 8, 812631. [Google Scholar] [CrossRef]
  62. Wang, S.; Konorev, E.A.; Kotamraju, S.; Joseph, J.; Kalivendi, S.; Kalyanaraman, B. Doxorubicin induces apoptosis in normal and tumor cells via distinctly different mechanisms. intermediacy of H(2)O(2)- and p53-dependent pathways. J. Biol. Chem. 2004, 279, 25535–25543. [Google Scholar] [CrossRef] [PubMed]
  63. Matsumoto, M.; Nakajima, W.; Seike, M.; Gemma, A.; Tanaka, N. Cisplatin-induced apoptosis in non-small-cell lung cancer cells is dependent on Bax- and Bak-induction pathway and synergistically activated by BH3-mimetic ABT-263 in p53 wild-type and mutant cells. Biochem. Biophys. Res. Commun. 2016, 473, 490–496. [Google Scholar] [CrossRef]
  64. Uchihara, Y.; Komori, R.; Tago, K.; Tamura, H.; Funakoshi-Tago, M. Methotrexate significantly induces apoptosis by inhibiting STAT3 activation in NPM-ALK-positive ALCL cells. Biochem. Pharmacol. 2019, 170, 113666. [Google Scholar] [CrossRef] [PubMed]
  65. Wong, R.S. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef]
  66. Bartlett, J.J.; Trivedi, P.C.; Pulinilkunnil, T. Autophagic dysregulation in doxorubicin cardiomyopathy. J. Mol. Cell. Cardiol. 2017, 104, 1–8. [Google Scholar] [CrossRef]
  67. Raja, W.; Mir, M.H.; Dar, I.; Banday, M.A.; Ahmad, I. Cisplatin induced paroxysmal supraventricular tachycardia. Indian J. Med. Paediatr. Oncol. 2013, 34, 330–332. [Google Scholar] [CrossRef] [PubMed]
  68. Zhu, H.; Sarkar, S.; Scott, L.; Danelisen, I.; Trush, M.A.; Jia, Z.; Li, Y.R. Doxorubicin Redox Biology: Redox Cycling, Topoisomerase Inhibition, and Oxidative Stress. React. Oxyg. Species 2016, 1, 189–198. [Google Scholar] [CrossRef]
  69. Yu, W.; Chen, Y.; Dubrulle, J.; Stossi, F.; Putluri, V.; Sreekumar, A.; Putluri, N.; Baluya, D.; Lai, S.Y.; Sandulache, V.C. Cisplatin generates oxidative stress which is accompanied by rapid shifts in central carbon metabolism. Sci. Rep. 2018, 8, 4306. [Google Scholar] [CrossRef]
  70. Hess, J.A.; Khasawneh, M.K. Cancer metabolism and oxidative stress: Insights into carcinogenesis and chemotherapy via the non-dihydrofolate reductase effects of methotrexate. BBA Clin. 2015, 3, 152–161. [Google Scholar] [CrossRef]
  71. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef] [PubMed]
  72. Russo, M.; Bono, E.; Ghigo, A. The Interplay Between Autophagy and Senescence in Anthracycline Cardiotoxicity. Curr. Heart Fail. Rep. 2021, 18, 180–190. [Google Scholar] [CrossRef]
  73. Chen, X.; Kang, R.; Kroemer, G.; Tang, D. Broadening horizons: The role of ferroptosis in cancer. Nat. Rev. Clin. Oncol. 2021, 18, 280–296. [Google Scholar] [CrossRef]
  74. Gong, Y.; Fan, Z.; Luo, G.; Yang, C.; Huang, Q.; Fan, K.; Cheng, H.; Jin, K.; Ni, Q.; Yu, X.; et al. The role of necroptosis in cancer biology and therapy. Mol. Cancer 2019, 18, 100. [Google Scholar] [CrossRef]
  75. Dong, Y.; Chen, H.; Gao, J.; Liu, Y.; Li, J.; Wang, J. Molecular machinery and interplay of apoptosis and autophagy in coronary heart disease. J. Mol. Cell. Cardiol. 2019, 136, 27–41. [Google Scholar] [CrossRef] [PubMed]
  76. Xie, L.H.; Fefelova, N.; Pamarthi, S.H.; Gwathmey, J.K. Molecular Mechanisms of Ferroptosis and Relevance to Cardiovascular Disease. Cells 2022, 11, 2726. [Google Scholar] [CrossRef]
  77. Dimitrakis, P.; Romay-Ogando, M.I.; Timolati, F.; Suter, T.M.; Zuppinger, C. Effects of doxorubicin cancer therapy on autophagy and the ubiquitin-proteasome system in long-term cultured adult rat cardiomyocytes. Cell Tissue. Res. 2012, 350, 361–372. [Google Scholar] [CrossRef] [PubMed]
  78. Li, D.L.; Hill, J.A. Cardiomyocyte autophagy and cancer chemotherapy. J. Mol. Cell. Cardiol. 2014, 71, 54–61. [Google Scholar] [CrossRef]
  79. Wang, F.; Chandra, J.; Kleinerman, E.S. Exercise intervention decreases acute and late doxorubicin-induced cardiotoxicity. Cancer Med. 2021, 10, 7572–7584. [Google Scholar] [CrossRef]
  80. Yang, X.; Li, G.; Yang, T.; Guan, M.; An, N.; Yang, F.; Dai, Q.; Zhong, C.; Luo, C.; Gao, Y.; et al. Possible Susceptibility Genes for Intervention against Chemotherapy-Induced Cardiotoxicity. Oxid. Med. Cell Longev. 2020, 2020, 4894625. [Google Scholar] [CrossRef]
  81. Schiattarella, G.G.; Hill, J.A. Therapeutic targeting of autophagy in cardiovascular disease. J. Mol. Cell. Cardiol. 2016, 95, 86–93. [Google Scholar] [CrossRef] [PubMed]
  82. Chun, Y.; Kim, J. Autophagy: An Essential Degradation Program for Cellular Homeostasis and Life. Cells 2018, 7, 278. [Google Scholar] [CrossRef] [PubMed]
  83. Lavandero, S.; Chiong, M.; Rothermel, B.A.; Hill, J.A. Autophagy in cardiovascular biology. J. Clin. Investig. 2015, 125, 55–64. [Google Scholar] [CrossRef] [PubMed]
  84. Swanson, K.V.; Deng, M.; Ting, J.P. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
  85. Wei, S.; Ma, W.; Li, X.; Jiang, C.; Sun, T.; Li, Y.; Zhang, B.; Li, W. Involvement of ROS/NLRP3 Inflammasome Signaling Pathway in Doxorubicin-Induced Cardiotoxicity. Cardiovasc. Toxicol. 2020, 20, 507–519. [Google Scholar] [CrossRef]
  86. Hua, Y.; Zhang, J.; Liu, Q.; Su, J.; Zhao, Y.; Zheng, G.; Yang, Z.; Zhuo, D.; Ma, C.; Fan, G. The Induction of Endothelial Autophagy and Its Role in the Development of Atherosclerosis. Front. Cardiovasc. Med. 2022, 9, 831847. [Google Scholar] [CrossRef]
  87. Schrijvers, D.M.; De Meyer, G.R.; Martinet, W. Autophagy in atherosclerosis: A potential drug target for plaque stabilization. Arter. Thromb. Vasc. Biol. 2011, 31, 2787–2791. [Google Scholar] [CrossRef]
  88. Singh, B.; Cui, K.; Eisa-Beygi, S.; Zhu, B.; Cowan, D.B.; Shi, J.; Wang, D.Z.; Liu, Z.; Bischoff, J.; Chen, H. Elucidating the crosstalk between endothelial-to-mesenchymal transition (EndoMT) and endothelial autophagy in the pathogenesis of atherosclerosis. Vasc. Pharmacol. 2024, 155, 107368. [Google Scholar] [CrossRef]
  89. Xu, A.; Deng, F.; Chen, Y.; Kong, Y.; Pan, L.; Liao, Q.; Rao, Z.; Xie, L.; Yao, C.; Li, S.; et al. NF-κB pathway activation during endothelial-to-mesenchymal transition in a rat model of doxorubicin-induced cardiotoxicity. Biomed. Pharmacother 2020, 130, 110525. [Google Scholar] [CrossRef]
  90. Cheng, Y.; Wang, Y.; Yin, R.; Xu, Y.; Zhang, L.; Zhang, Y.; Yang, L.; Zhao, D. Central role of cardiac fibroblasts in myocardial fibrosis of diabetic cardiomyopathy. Front. Endocrinol. 2023, 14, 1162754. [Google Scholar] [CrossRef]
  91. Curigliano, G.; Lenihan, D.; Fradley, M.; Ganatra, S.; Barac, A.; Blaes, A.; Herrmann, J.; Porter, C.; Lyon, A.R.; Lancellotti, P.; et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann. Oncol. 2020, 31, 171–190. [Google Scholar] [CrossRef] [PubMed]
  92. Hu, Y.; Zhou, Y.; Dai, N.; Song, S.; Zhao, X.; Zhao, Y.; Cheng, L.; Lu, H.; Ge, J. Enhancing panvascular medicine: Unveiling the nexus of pan-cardio-oncology and expanding therapeutic frontiers. Sci. Bull. 2025, 70, 798–800. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, D.; Peyre, F.; Loissell-Baltazar, Y.A.; Courilleau, D.; Lacas-Gervais, S.; Nicolas, V.; Jacquet, E.; Dokudovskaya, S.; Taran, F.; Cintrat, J.C.; et al. Identification of Small Molecules Inhibiting Cardiomyocyte Necrosis and Apoptosis by Autophagy Induction and Metabolism Reprogramming. Cells 2022, 11, 474. [Google Scholar] [CrossRef]
  94. Turcotte, S.; Chan, D.A.; Sutphin, P.D.; Hay, M.P.; Denny, W.A.; Giaccia, A.J. A molecule targeting VHL-deficient renal cell carcinoma that induces autophagy. Cancer Cell 2008, 14, 90–102. [Google Scholar] [CrossRef]
  95. Gao, G.; Chen, W.; Yan, M.; Liu, J.; Luo, H.; Wang, C.; Yang, P. Rapamycin regulates the balance between cardiomyocyte apoptosis and autophagy in chronic heart failure by inhibiting mTOR signaling. Int. J. Mol. Med. 2020, 45, 195–209. [Google Scholar] [CrossRef] [PubMed]
  96. Li, J.; Kim, S.G.; Blenis, J. Rapamycin: One drug, many effects. Cell Metab. 2014, 19, 373–379. [Google Scholar] [CrossRef]
  97. Shoji-Kawata, S.; Sumpter, R.; Leveno, M.; Campbell, G.R.; Zou, Z.; Kinch, L.; Wilkins, A.D.; Sun, Q.; Pallauf, K.; MacDuff, D.; et al. Identification of a candidate therapeutic autophagy-inducing peptide. Nature 2013, 494, 201–206. [Google Scholar] [CrossRef]
  98. Peraro, L.; Zou, Z.; Makwana, K.M.; Cummings, A.E.; Ball, H.L.; Yu, H.; Lin, Y.S.; Levine, B.; Kritzer, J.A. Diversity-Oriented Stapling Yields Intrinsically Cell-Penetrant Inducers of Autophagy. J. Am. Chem. Soc. 2017, 139, 7792–7802. [Google Scholar] [CrossRef]
  99. Zou, Y.; Spokoyny, A.M.; Zhang, C.; Simon, M.D.; Yu, H.; Lin, Y.S.; Pentelute, B.L. Convergent diversity-oriented side-chain macrocyclization scan for unprotected polypeptides. Org. Biomol. Chem. 2014, 12, 566–573. [Google Scholar] [CrossRef]
  100. Zagkou, S.; Marais, V.; Zeghoudi, N.; Guillou, E.L.; Eskelinen, E.L.; Panasyuk, G.; Verrier, B.; Primard, C. Design and Evaluation of Autophagy-Inducing Particles for the Treatment of Abnormal Lipid Accumulation. Pharmaceutics 2022, 14, 1379. [Google Scholar] [CrossRef]
  101. Zhou, Z.; Yan, Y.; Wang, L.; Zhang, Q.; Cheng, Y. Melanin-like nanoparticles decorated with an autophagy-inducing peptide for efficient targeted photothermal therapy. Biomaterials 2019, 203, 63–72. [Google Scholar] [CrossRef]
  102. Zahid, M.; Feldman, K.S.; Garcia-Borrero, G.; Feinstein, T.N.; Pogodzinski, N.; Xu, X.; Yurko, R.; Czachowski, M.; Wu, Y.L.; Mason, N.S.; et al. Cardiac Targeting Peptide, a Novel Cardiac Vector: Studies in Bio-Distribution, Imaging Application, and Mechanism of Transduction. Biomolecules 2018, 8, 147. [Google Scholar] [CrossRef] [PubMed]
  103. Elmorshdy Elsaeed Mohammed Elmorshdy, S.; Ahmed Shaker, G.; Helmy Eldken, Z.; Abdelbadie Salem, M.; Awadalla, A.; Mahmoud Abdel Shakour, H.; Elmahdy El Hosiny Sarhan, M.; Mohamed Hussein, A. Impact of Cerium Oxide Nanoparticles on Metabolic, Apoptotic, Autophagic and Antioxidant Changes in Doxorubicin-Induced Cardiomyopathy: Possible Underlying Mechanisms. Rep. Biochem. Mol. Biol. 2023, 12, 495–511. [Google Scholar] [CrossRef]
  104. Taheriazam, A.; Abad, G.G.Y.; Hajimazdarany, S.; Imani, M.H.; Ziaolhagh, S.; Zandieh, M.A.; Bayanzadeh, S.D.; Mirzaei, S.; Hamblin, M.R.; Entezari, M.; et al. Graphene oxide nanoarchitectures in cancer biology: Nano-modulators of autophagy and apoptosis. J. Control. Release 2023, 354, 503–522. [Google Scholar] [CrossRef] [PubMed]
  105. Khan, A.; Bakhru, P.; Saikolappan, S.; Das, K.; Soudani, E.; Singh, C.R.; Estrella, J.L.; Zhang, D.; Pasare, C.; Ma, Y.; et al. An autophagy-inducing and TLR-2 activating BCG vaccine induces a robust protection against tuberculosis in mice. NPJ Vaccines 2019, 4, 34. [Google Scholar] [CrossRef] [PubMed]
  106. Alsaleh, G.; Panse, I.; Swadling, L.; Zhang, H.; Richter, F.C.; Meyer, A.; Lord, J.; Barnes, E.; Klenerman, P.; Green, C.; et al. Autophagy in T cells from aged donors is maintained by spermidine and correlates with function and vaccine responses. eLife 2020, 9, e57950. [Google Scholar] [CrossRef]
  107. Clancy, J.W.; D’Souza-Schorey, C. Tumor-Derived Extracellular Vesicles: Multifunctional Entities in the Tumor Microenvironment. Annu. Rev. Pathol. 2023, 18, 205–229. [Google Scholar] [CrossRef]
  108. Kalluri, R. The biology and function of exosomes in cancer. J. Clin. Investig. 2016, 126, 1208–1215. [Google Scholar] [CrossRef]
  109. Kanada, M.; Bachmann, M.H.; Contag, C.H. Signaling by Extracellular Vesicles Advances Cancer Hallmarks. Trends Cancer. 2016, 2, 84–94. [Google Scholar] [CrossRef]
  110. Kumar, M.A.; Baba, S.K.; Sadida, H.Q.; Marzooqi, S.A.; Jerobin, J.; Altemani, F.H.; Algehainy, N.; Alanazi, M.A.; Abou-Samra, A.B.; Kumar, R.; et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct. Target. Ther. 2024, 9, 27. [Google Scholar]
  111. Li, Z.; Guo, K.; Gao, Z.; Chen, J.; Ye, Z.; Cao, M.; Wang, S.E.; Yin, Y.; Zhong, W. Colocalization of protein and microRNA markers reveals unique extracellular vesicle subpopulations for early cancer detection. Sci. Adv. 2024, 10, eadh8689. [Google Scholar] [CrossRef] [PubMed]
  112. Zhou, E.; Li, Y.; Wu, F.; Guo, M.; Xu, J.; Wang, S.; Tan, Q.; Ma, P.; Song, S.; Jin, Y. Circulating extracellular vesicles are effective biomarkers for predicting response to cancer therapy. eBioMedicine 2021, 67, 103365. [Google Scholar] [CrossRef]
  113. Beaumier, A.; Robinson, S.R.; Robinson, N.; Lopez, K.E.; Meola, D.M.; Barber, L.G.; Bulmer, B.J.; Calvalido, J.; Rush, J.E.; Yeri, A.; et al. Extracellular vesicular microRNAs as potential biomarker for early detection of doxorubicin-induced cardiotoxicity. J. Vet. Intern. Med. 2020, 34, 1260–1271. [Google Scholar] [CrossRef] [PubMed]
  114. Boen, H.M.; Cherubin, M.; Franssen, C.; Gevaert, A.B.; Witvrouwen, I.; Bosman, M.; Guns, P.-J.; Heidbuchel, H.; Loeys, B.; Alaerts, M.; et al. Circulating MicroRNA as Biomarkers of Anthracycline-Induced Cardiotoxicity: JACC: CardioOncology State-of-the-Art Review. JACC Cardio. Oncol. 2024, 6, 183–199. [Google Scholar] [CrossRef]
  115. Milano, G.; Biemmi, V.; Lazzarini, E.; Balbi, C.; Ciullo, A.; Bolis, S.; Ameri, P.; Di Silvestre, D.; Mauri, P.; Barile, L.; et al. Intravenous administration of cardiac progenitor cell-derived exosomes protects against doxorubicin/trastuzumab-induced cardiac toxicity. Cardiovasc. Res. 2020, 116, 383–392. [Google Scholar] [CrossRef]
  116. Tian, C.; Yang, Y.; Bai, B.; Wang, S.; Liu, M.; Sun, R.C.; Yu, T.; Chu, X.M. Potential of exosomes as diagnostic biomarkers and therapeutic carriers for doxorubicin-induced cardiotoxicity. Int. J. Biol. Sci. 2021, 17, 1328–1338. [Google Scholar] [CrossRef] [PubMed]
  117. Yarana, C.; Carroll, D.; Chen, J.; Chaiswing, L.; Zhao, Y.; Noel, T.; Alstott, M.; Bae, Y.; Dressler, E.V.; Moscow, J.A.; et al. Extracellular Vesicles Released by Cardiomyocytes in a Doxorubicin-Induced Cardiac Injury Mouse Model Contain Protein Biomarkers of Early Cardiac Injury. Clin. Cancer Res. 2018, 24, 1644–1653. [Google Scholar] [CrossRef]
  118. Luo, R.; Liu, M.; Tan, T.; Yang, Q.; Wang, Y.; Men, L.; Zhao, L.; Zhang, H.; Wang, S.; Xie, T.; et al. Emerging Significance and Therapeutic Potential of Extracellular vesicles. Int. J. Biol. Sci. 2021, 17, 2476–2486. [Google Scholar] [CrossRef]
  119. Murphy, D.E.; de Jong, O.G.; Brouwer, M.; Wood, M.J.; Lavieu, G.; Schiffelers, R.M.; Vader, P. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp. Mol. Med. 2019, 51, 1–12. [Google Scholar] [CrossRef]
  120. Sun, M.; Yang, J.; Fan, Y.; Zhang, Y.; Sun, J.; Hu, M.; Sun, K.; Zhang, J. Beyond Extracellular Vesicles: Hybrid Membrane Nanovesicles as Emerging Advanced Tools for Biomedical Applications. Adv. Sci. 2023, 10, e2303617. [Google Scholar] [CrossRef]
  121. Wiklander, O.P.B.; Brennan, M.A.; Lotvall, J.; Breakefield, X.O.; El Andaloussi, S. Advances in therapeutic applications of extracellular vesicles. Sci. Transl. Med. 2019, 11, eaav8521. [Google Scholar] [CrossRef] [PubMed]
  122. Yang, L.; Patel, K.D.; Rathnam, C.; Thangam, R.; Hou, Y.; Kang, H.; Lee, K.B. Harnessing the Therapeutic Potential of Extracellular Vesicles for Biomedical Applications Using Multifunctional Magnetic Nanomaterials. Small 2022, 18, e2104783. [Google Scholar] [CrossRef] [PubMed]
  123. Yong, T.; Zhang, X.; Bie, N.; Zhang, H.; Zhang, X.; Li, F.; Hakeem, A.; Hu, J.; Gan, L.; Santos, H.A.; et al. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun. 2019, 10, 3838. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, X.; Zhang, H.; Gu, J.; Zhang, J.; Shi, H.; Qian, H.; Wang, D.; Xu, W.; Pan, J.; Santos, H.A. Engineered Extracellular Vesicles for Cancer Therapy. Adv. Mater. 2021, 33, e2005709. [Google Scholar] [CrossRef]
  125. Colombo, M.; Raposo, G.; Thery, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  126. Liu, J.; Jiang, M.; Deng, S.; Lu, J.; Huang, H.; Zhang, Y.; Gong, P.; Shen, X.; Ruan, H.; Jin, M.; et al. miR-93-5p-Containing Exosomes Treatment Attenuates Acute Myocardial Infarction-Induced Myocardial Damage. Mol Ther. Nucleic. Acids. 2018, 11, 103–115. [Google Scholar] [CrossRef]
  127. Wei, W.; Pan, Y.; Yang, X.; Chen, Z.; Heng, Y.; Yang, B.; Pu, M.; Zuo, J.; Lai, Z.; Tang, Y.; et al. The Emerging Role of the Interaction of Extracellular Vesicle and Autophagy-Novel Insights into Neurological Disorders. J. Inflamm. Res. 2022, 15, 3395–3407. [Google Scholar] [CrossRef]
  128. Santoso, M.R.; Ikeda, G.; Tada, Y.; Jung, J.H.; Vaskova, E.; Sierra, R.G.; Gati, C.; Goldstone, A.B.; von Bornstaedt, D.; Shukla, P.; et al. Exosomes From Induced Pluripotent Stem Cell-Derived Cardiomyocytes Promote Autophagy for Myocardial Repair. J. Am. Heart Assoc. 2020, 9, e014345. [Google Scholar] [CrossRef]
  129. Zhao, S.; Liu, Y.; Pu, Z. Bone marrow mesenchymal stem cell-derived exosomes attenuate D-GaIN/LPS-induced hepatocyte apoptosis by activating autophagy in vitro. Drug Des. Dev. Ther. 2019, 13, 2887–2897. [Google Scholar] [CrossRef]
  130. Rong, Y.; Liu, W.; Wang, J.; Fan, J.; Luo, Y.; Li, L.; Kong, F.; Chen, J.; Tang, P.; Cai, W. Neural stem cell-derived small extracellular vesicles attenuate apoptosis and neuroinflammation after traumatic spinal cord injury by activating autophagy. Cell Death Dis. 2019, 10, 340. [Google Scholar] [CrossRef]
  131. Jiang, X.; Lew, K.S.; Chen, Q.; Richards, A.M.; Wang, P. Human Mesenchymal Stem Cell-derived Exosomes Reduce Ischemia/Reperfusion Injury by the Inhibitions of Apoptosis and Autophagy. Curr. Pharm. Des. 2018, 24, 5334–5341. [Google Scholar] [CrossRef]
  132. Keulers, T.G.; Libregts, S.F.; Beaumont, J.E.J.; Savelkouls, K.G.; Bussink, J.; Duimel, H.; Dubois, L.; Zonneveld, M.I.; Lopez-Iglesias, C.; Bezstarosti, K.; et al. Secretion of pro-angiogenic extracellular vesicles during hypoxia is dependent on the autophagy-related protein GABARAPL. J. Extracell Vesicles. 2021, 10, e12166. [Google Scholar] [CrossRef]
  133. Leidal, A.M.; Huang, H.H.; Marsh, T.; Solvik, T.; Zhang, D.; Ye, J.; Kai, F.; Goldsmith, J.; Liu, J.Y.; Huang, Y.H.; et al. The LC3-conjugation machinery specifies the loading of RNA-binding proteins into extracellular vesicles. Nat. Cell Biol. 2020, 22, 187–199. [Google Scholar] [CrossRef] [PubMed]
  134. Solvik, T.A.; Nguyen, T.A.; Tony Lin, Y.H.; Marsh, T.; Huang, E.J.; Wiita, A.P.; Debnath, J.; Leidal, A.M. Secretory autophagy maintains proteostasis upon lysosome inhibition. J. Cell Biol. 2022, 221, e202110151. [Google Scholar] [CrossRef] [PubMed]
  135. Xu, X.; Xia, T. Recent Advances in Site-Specific Lipid Nanoparticles for mRNA Delivery. ACS Nanosci Au. 2023, 3, 192–203. [Google Scholar] [CrossRef]
  136. Lin, Y.X.; Wang, Y.; Ding, J.; Jiang, A.; Wang, J.; Yu, M.; Blake, S.; Liu, S.; Bieberich, C.J.; Farokhzad, O.C.; et al. Reactivation of the tumor suppressor PTEN by mRNA nanoparticles enhances antitumor immunity in preclinical models. Sci. Transl. Med. 2021, 13, eaba9772. [Google Scholar] [CrossRef] [PubMed]
  137. Brown, H.; Chung, M.; Uffing, A.; Batistatou, N.; Tsang, T.; Doskocil, S.; Mao, W.; Willbold, D.; Bast, R.C., Jr.; Lu, Z.; et al. Structure-Based Design of Stapled Peptides That Bind GABARAP and Inhibit Autophagy. J. Am. Chem. Soc. 2022, 144, 14687–14697. [Google Scholar] [CrossRef]
  138. Njomen, E.; Tepe, J.J. Regulation of Autophagic Flux by the 20S Proteasome. Cell Chem. Biol. 2019, 26, 1283–1294.e5. [Google Scholar] [CrossRef]
  139. Jiang, Z.; Kuo, Y.H.; Arkin, M.R. Autophagy Receptor-Inspired Antibody-Fusion Proteins for Targeted Intracellular Degradation. J. Am. Chem. Soc. 2023, 145, 23939–23947. [Google Scholar] [CrossRef]
  140. Ji, C.H.; Kim, H.Y.; Lee, M.J.; Heo, A.J.; Park, D.Y.; Lim, S.; Shin, S.; Ganipisetti, S.; Yang, W.S.; Jung, C.A.; et al. The AUTOTAC chemical biology platform for targeted protein degradation via the autophagy-lysosome system. Nat. Commun. 2022, 13, 904. [Google Scholar] [CrossRef]
  141. Xie, C.; Zhuang, X.X.; Niu, Z.; Ai, R.; Lautrup, S.; Zheng, S.; Jiang, Y.; Han, R.; Gupta, T.S.; Cao, S.; et al. Amelioration of Alzheimer’s disease pathology by mitophagy inducers identified via machine learning and a cross-species workflow. Nat. Biomed. Eng. 2022, 6, 76–93. [Google Scholar] [CrossRef] [PubMed]
  142. Kapuy, O.; Papp, D.; Vellai, T.; Bánhegyi, G.; Korcsmáros, T. Systems-Level Feedbacks of NRF2 Controlling Autophagy upon Oxidative Stress Response. Antioxidants 2018, 7, 39. [Google Scholar] [CrossRef]
  143. Shirin, A.; Klickstein, I.S.; Feng, S.; Lin, Y.T.; Hlavacek, W.S.; Sorrentino, F. Prediction of Optimal Drug Schedules for Controlling Autophagy. Sci. Rep. 2019, 9, 1428. [Google Scholar] [CrossRef] [PubMed]
  144. Burridge, P.W.; Li, Y.F.; Matsa, E.; Wu, H.; Ong, S.G.; Sharma, A.; Holmstrom, A.; Chang, A.C.; Coronado, M.J.; Ebert, A.D.; et al. Human induced pluripotent stem cell-derived cardiomyocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat. Med. 2016, 22, 547–556. [Google Scholar] [CrossRef] [PubMed]
  145. Cao, X.; Thomas, D.; Whitcomb, L.A.; Wang, M.; Chatterjee, A.; Chicco, A.J.; Weil, M.M.; Wu, J.C. Modeling ionizing radiation-induced cardiovascular dysfunction with human iPSC-derived engineered heart tissues. J. Mol. Cell. Cardiol. 2024, 188, 105–107. [Google Scholar] [CrossRef]
  146. Stack, J.P.; Moslehi, J.; Sayed, N.; Wu, J.C. Cancer therapy-induced cardiomyopathy: Can human induced pluripotent stem cell modelling help prevent it? Eur. Heart J. 2019, 40, 1764–1770. [Google Scholar] [CrossRef]
  147. Boukhalfa, A.; Robinson, S.R.; Meola, D.M.; Robinson, N.A.; Ling, L.A.; LaMastro, J.N.; Upshaw, J.N.; Pulakat, L.; Jaffe, I.Z.; London, C.A.; et al. Using cultured canine cardiac slices to model the autophagic flux with doxorubicin. PLoS ONE 2023, 18, e0282859. [Google Scholar] [CrossRef]
  148. Watson, S.A.; Scigliano, M.; Bardi, I.; Ascione, R.; Terracciano, C.M.; Perbellini, F. Preparation of viable adult ventricular myocardial slices from large and small mammals. Nat. Protoc. 2017, 12, 2623–2639. [Google Scholar] [CrossRef]
  149. Kang, C.; Qiao, Y.; Li, G.; Baechle, K.; Camelliti, P.; Rentschler, S.; Efimov, I.R. Human Organotypic Cultured Cardiac Slices: New Platform For High Throughput Preclinical Human Trials. Sci. Rep. 2016, 6, 28798. [Google Scholar] [CrossRef]
  150. Perbellini, F.; Thum, T. Living myocardial slices: A novel multicellular model for cardiac translational research. Eur. Heart J. 2020, 41, 2405–2408. [Google Scholar] [CrossRef]
  151. Ross, A.J.; Krumova, I.; Tunc, B.; Wu, Q.; Wu, C.; Camelliti, P. A novel method to extend viability and functionality of living heart slices. Front. Cardiovasc. Med. 2023, 10, 1244630. [Google Scholar] [CrossRef] [PubMed]
  152. Lehman, H.L.; Stairs, D.B. Single and Multiple Gene Manipulations in Mouse Models of Human Cancer. Cancer Growth Metastasis 2015, 8, 1–15. [Google Scholar] [CrossRef]
  153. Olson, B.; Li, Y.; Lin, Y.; Liu, E.T.; Patnaik, A. Mouse Models for Cancer Immunotherapy Research. Cancer Discov. 2018, 8, 1358–1365. [Google Scholar] [CrossRef]
  154. Asnani, A.; Moslehi, J.J.; Adhikari, B.B.; Baik, A.H.; Beyer, A.M.; de Boer, R.A.; Ghigo, A.; Grumbach, I.M.; Jain, S.; Zhu, H.; et al. Preclinical Models of Cancer Therapy-Associated Cardiovascular Toxicity: A Scientific Statement From the American Heart Association. Circ. Res. 2021, 129, e21–e34. [Google Scholar] [CrossRef] [PubMed]
  155. Salloum, F.N.; Tocchetti, C.G.; Ameri, P.; Ardehali, H.; Asnani, A.; de Boer, R.A.; Burridge, P.; Cabrera, J.A.; de Castro, J.; Cordoba, R.; et al. Priorities in Cardio-Oncology Basic and Translational Science: GCOS 2023 Symposium Proceedings: JACC: CardioOncology State-of-the-Art Review. JACC Cardio Oncol. 2023, 5, 715–731. [Google Scholar] [CrossRef] [PubMed]
  156. Hallman, B.E.; Hauck, M.L.; Williams, L.E.; Hess, P.R.; Suter, S.E. Incidence and risk factors associated with development of clinical cardiotoxicity in dogs receiving doxorubicin. J. Vet. Int. Med. 2019, 33, 783–791. [Google Scholar] [CrossRef]
  157. Tjostheim, S.S.; Stepien, R.L.; Markovic, L.E.; Stein, T.J. Effects of Toceranib Phosphate on Systolic Blood Pressure and Proteinuria in Dogs. J. Vet. Int. Med. 2016, 30, 951–957. [Google Scholar] [CrossRef]
  158. Zhao, Y.G.; Codogno, P.; Zhang, H. Machinery, regulation and pathophysiological implications of autophagosome maturation. Nat. Rev. Mol. Cell Biol. 2021, 22, 733–750. [Google Scholar] [CrossRef]
  159. Beesabathuni, N.S.; Park, S.; Shah, P.S. Quantitative and temporal measurement of dynamic autophagy rates. Autophagy 2023, 19, 1164–1183. [Google Scholar] [CrossRef]
  160. Kabeya, Y.; Mizushima, N.; Ueno, T.; Yamamoto, A.; Kirisako, T.; Noda, T.; Kominami, E.; Ohsumi, Y.; Yoshimori, T. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000, 19, 5720–5728. [Google Scholar] [CrossRef]
  161. Kaizuka, T.; Morishita, H.; Hama, Y.; Tsukamoto, S.; Matsui, T.; Toyota, Y.; Kodama, A.; Ishihara, T.; Mizushima, T.; Mizushima, N. An Autophagic Flux Probe that Releases an Internal Control. Mol. Cell 2016, 64, 835–849. [Google Scholar] [CrossRef] [PubMed]
  162. Mizushima, N.; Yamamoto, A.; Matsui, M.; Yoshimori, T.; Ohsumi, Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol. Biol. Cell 2004, 15, 1101–1111. [Google Scholar] [CrossRef] [PubMed]
  163. Moulis, M.; Vindis, C. Methods for Measuring Autophagy in Mice. Cells 2017, 6, 14. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, H.H.; Khatun, Z.; Wei, L.; Mekkaoui, C.; Patel, D.; Kim, S.J.W.; Boukhalfa, A.; Enoma, E.; Meng, L.; Chen, Y.I.; et al. A nanoparticle probe for the imaging of autophagic flux in live mice via magnetic resonance and near-infrared fluorescence. Nat. Biomed. Eng. 2022, 6, 1045–1056. [Google Scholar] [CrossRef]
  165. Toth, G.B.; Varallyay, C.G.; Horvath, A.; Bashir, M.R.; Choyke, P.L.; Daldrup-Link, H.E.; Dosa, E.; Finn, J.P.; Gahramanov, S.; Harisinghani, M.; et al. Current and potential imaging applications of ferumoxytol for magnetic resonance imaging. Kidney Int. 2017, 92, 47–66. [Google Scholar] [CrossRef]
  166. Bhatia, S.N.; Chen, X.; Dobrovolskaia, M.A.; Lammers, T. Cancer nanomedicine. Nat. Rev. Cancer 2022, 22, 550–556. [Google Scholar] [CrossRef]
  167. Chehelgerdi, M.; Chehelgerdi, M.; Allela, O.Q.B.; Pecho, R.D.C.; Jayasankar, N.; Rao, D.P.; Thamaraikani, T.; Vasanthan, M.; Viktor, P.; Lakshmaiya, N.; et al. Progressing nanotechnology to improve targeted cancer treatment: Overcoming hurdles in its clinical implementation. Mol. Cancer 2023, 22, 169. [Google Scholar] [CrossRef]
  168. Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
  169. Borges, G.S.M.; Lages, E.B.; Sicard, P.; Ferreira, L.A.M.; Richard, S. Nanomedicine in Oncocardiology: Contribution and Perspectives of Preclinical Studies. Front. Cardiovasc. Med. 2021, 8, 690533. [Google Scholar] [CrossRef]
  170. Nevins, S.; McLoughlin, C.D.; Oliveros, A.; Stein, J.B.; Rashid, M.A.; Hou, Y.; Jang, M.H.; Lee, K.B. Nanotechnology Approaches for Prevention and Treatment of Chemotherapy-Induced Neurotoxicity, Neuropathy, and Cardiomyopathy in Breast and Ovarian Cancer Survivors. Small 2023, 20, e2300744. [Google Scholar] [CrossRef]
  171. Smith, B.R.; Edelman, E.R. Nanomedicines for cardiovascular disease. Nat. Cardiovasc. Res. 2023, 2, 351–367. [Google Scholar] [CrossRef] [PubMed]
  172. Shan, C.; Chen, X.; Cai, H.; Hao, X.; Li, J.; Zhang, Y.; Gao, J.; Zhou, Z.; Li, X.; Liu, C.; et al. The Emerging Roles of Autophagy-Related MicroRNAs in Cancer. Int. J. Biol. Sci. 2021, 17, 134–150. [Google Scholar] [CrossRef] [PubMed]
  173. Luo, M.; Ye, L.; Chang, R.; Ye, Y.; Zhang, Z.; Liu, C.; Li, S.; Jing, Y.; Ruan, H.; Zhang, G.; et al. Multi-omics characterization of autophagy-related molecular features for therapeutic targeting of autophagy. Nat. Commun. 2022, 13, 6345. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Autophagy modulations by genetic, pharmacological, or dietary means target specific steps of the autophagic pathway. Autophagy can be induced genetically by inhibitors of the mTORC1 and mTORC2 complexes, the activation of the AMP kinase pathway, and the downstream ULK1 and PI3K complexes. Pharmacological interventions such as rapamycin [14,15], everolimus [16], and torin1 [17] are robust mTOR inhibitors and activate autophagy. Dietary intervention by starvation [18] or pharmacologic metformin [19] induces autophagy via the mTOR and AMPK pathways. Trehalose inhibits glucose uptake, inducing starvation [20]. Schizandrin A induces autophagy and apoptosis in cancer cells [21] but suppresses autophagy during neuronal injury [22]. Downstream in the autophagic process, lysosomotropic agents such as chloroquine prevent autophagosome and lysosome fusion [23] and inhibit autophagy. Bafilomycin A1 inhibits autophagy by targeting the vacuolar-type H-ATPase (V-ATPase) proton pumps on lysosomes [24]. Tomatidine promotes lysosomal activity via the activation of transcription factor EB (TFEB) [25]. Autophagy modulation and inhibition differentially impact cell death and viability in the heart and in tumors.
Figure 1. Autophagy modulations by genetic, pharmacological, or dietary means target specific steps of the autophagic pathway. Autophagy can be induced genetically by inhibitors of the mTORC1 and mTORC2 complexes, the activation of the AMP kinase pathway, and the downstream ULK1 and PI3K complexes. Pharmacological interventions such as rapamycin [14,15], everolimus [16], and torin1 [17] are robust mTOR inhibitors and activate autophagy. Dietary intervention by starvation [18] or pharmacologic metformin [19] induces autophagy via the mTOR and AMPK pathways. Trehalose inhibits glucose uptake, inducing starvation [20]. Schizandrin A induces autophagy and apoptosis in cancer cells [21] but suppresses autophagy during neuronal injury [22]. Downstream in the autophagic process, lysosomotropic agents such as chloroquine prevent autophagosome and lysosome fusion [23] and inhibit autophagy. Bafilomycin A1 inhibits autophagy by targeting the vacuolar-type H-ATPase (V-ATPase) proton pumps on lysosomes [24]. Tomatidine promotes lysosomal activity via the activation of transcription factor EB (TFEB) [25]. Autophagy modulation and inhibition differentially impact cell death and viability in the heart and in tumors.
Pharmaceuticals 18 00671 g001
Figure 2. Cross-links between autophagy modulators, cancer treatments, cancer, and the heart. Current clinical practices in oncology and cardio-oncology focus on the interplay between cancer, its treatments, and their long-term effects on cardiac function. While autophagy has been shown to have promising cardioprotective effects in certain contexts, there is currently a lack of understanding regarding its effects on different cell types, especially under various disease and treatment conditions. Further investigations of autophagy’s mechanism and kinetics, as well as the creation of a translational cardio-oncology model, are required to realize its potential as a therapeutic strategy.
Figure 2. Cross-links between autophagy modulators, cancer treatments, cancer, and the heart. Current clinical practices in oncology and cardio-oncology focus on the interplay between cancer, its treatments, and their long-term effects on cardiac function. While autophagy has been shown to have promising cardioprotective effects in certain contexts, there is currently a lack of understanding regarding its effects on different cell types, especially under various disease and treatment conditions. Further investigations of autophagy’s mechanism and kinetics, as well as the creation of a translational cardio-oncology model, are required to realize its potential as a therapeutic strategy.
Pharmaceuticals 18 00671 g002
Figure 3. Overcoming the translational hurdles of autophagy modulation in cardio-oncology. The effects of chemotherapeutics, autophagy response to cancer drugs, and autophagy modulation must be investigated simultaneously in the heart and tumors. Various interdisciplinary approaches can be taken, including cell biology, 3D ex vivo tissue models, and in vivo preclinical mouse and translational canine cancer models. Specifically, pet dogs that spontaneously develop cancer and receive subsequent cancer therapy can develop cardiovascular dysfunction, thus serving as a uniquely valuable translational model. The cross-species platform, combined with diagnostic and therapeutic nanotechnology, can successfully impact the clinical management of millions of cancer patients and survivors who face an increased yet unpredictable risk of developing cardiovascular dysfunction, reducing unwanted cardiotoxicity, enhancing cancer treatment efficacy, and improving clinical outcomes.
Figure 3. Overcoming the translational hurdles of autophagy modulation in cardio-oncology. The effects of chemotherapeutics, autophagy response to cancer drugs, and autophagy modulation must be investigated simultaneously in the heart and tumors. Various interdisciplinary approaches can be taken, including cell biology, 3D ex vivo tissue models, and in vivo preclinical mouse and translational canine cancer models. Specifically, pet dogs that spontaneously develop cancer and receive subsequent cancer therapy can develop cardiovascular dysfunction, thus serving as a uniquely valuable translational model. The cross-species platform, combined with diagnostic and therapeutic nanotechnology, can successfully impact the clinical management of millions of cancer patients and survivors who face an increased yet unpredictable risk of developing cardiovascular dysfunction, reducing unwanted cardiotoxicity, enhancing cancer treatment efficacy, and improving clinical outcomes.
Pharmaceuticals 18 00671 g003
Table 1. Differential effects of cancer drugs on autophagy and associated processes in the heart and tumors.
Table 1. Differential effects of cancer drugs on autophagy and associated processes in the heart and tumors.
Biological EffectsCancer Drugs’ Impact on the HeartCancer Drugs’ Impact on TumorsBiological Effects Seen in Untreated Tumors
Autophagy
  • Reduced by doxorubicin [51]
  • Induced by cisplatin [52]
  • Reduced by doxorubicin [53]
  • Induced by cisplatin [54]
  • Reduced during tumor initiation; induced during malignancy [36,37]
Autosis
  • Induced by doxorubicin [55]
  • Induced by cisplatin [56]
  • Induced by methotrexate [57]
  • Induced with increased autophagy levels [58]
Apoptosis
  • Induced by doxorubicin [59]
  • Induced by cisplatin [60]
  • Induced by methotrexate [61]
  • Induced by doxorubicin [62]
  • Induced by cisplatin [63]
  • Induced by methotrexate [64]
  • Reduced in tumors [65]
Oxidative Stress
  • Induced by doxorubicin [66]
  • Induced by cisplatin [67]
  • Induced by doxorubicin [68]
  • Induced by cisplatin [69]
  • Induced by methotrexate [70]
  • Induced in tumors [71]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ling, L.A.; Boukhalfa, A.; Kung, A.H.; Yang, V.K.; Chen, H.H. Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity. Pharmaceuticals 2025, 18, 671. https://doi.org/10.3390/ph18050671

AMA Style

Ling LA, Boukhalfa A, Kung AH, Yang VK, Chen HH. Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity. Pharmaceuticals. 2025; 18(5):671. https://doi.org/10.3390/ph18050671

Chicago/Turabian Style

Ling, Lauren A., Asma Boukhalfa, Andrew H. Kung, Vicky K. Yang, and Howard H. Chen. 2025. "Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity" Pharmaceuticals 18, no. 5: 671. https://doi.org/10.3390/ph18050671

APA Style

Ling, L. A., Boukhalfa, A., Kung, A. H., Yang, V. K., & Chen, H. H. (2025). Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity. Pharmaceuticals, 18(5), 671. https://doi.org/10.3390/ph18050671

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