Next Article in Journal
Living Bacteriophage Engineering for Functional Material Synthesis, Bioanalytical Sensing and Disease Theranostics
Previous Article in Journal
Unit-Emitting Carbon Dots for Cell Imaging and Lipid Droplet Quantification
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Positive Inotropic Agents in Cancer Therapy: Exploring Potential Anti-Tumor Effects

1
PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
2
CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
3
ICBAS—School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
4
Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
*
Author to whom correspondence should be addressed.
Targets 2024, 2(2), 137-156; https://doi.org/10.3390/targets2020009
Submission received: 26 April 2024 / Revised: 28 May 2024 / Accepted: 7 June 2024 / Published: 13 June 2024

Abstract

:
Cancer remains a significant global health challenge despite advancements in diagnosis and treatment. Traditional cancer therapies often face limitations such as toxicity and drug resistance. Drug repurposing has emerged as a promising strategy to overcome these challenges by identifying new therapeutic uses for existing drugs. This review explores the potential of repurposing positive inotropic agents, which are traditionally used in cardiovascular medicine, for cancer therapy. Positive inotropic agents, including cardiac glycosides, β-agonists, phosphodiesterase inhibitors, and calcium sensitizers have shown preclinical evidence of anti-tumor activity through various mechanisms, such as modulation of the intracellular signaling pathways, increasing cyclic adenosine monophosphate (cAMP) levels, the production of nitric oxide, and decreasing reactive oxygen species levels. Despite the absence of specific clinical trials in this area, these findings suggest a promising avenue for further research and development of combination therapies to improve cancer treatment outcomes. However, challenges such as elucidating specific anti-tumor mechanisms, identifying predictive biomarkers, and optimizing safety profiles need to be addressed to fully realize the therapeutic potential of positive inotropic agents in oncology.

1. Introduction

Cancer is an extensive and heterogeneous disease that continues to challenge the global healthcare landscape. Characterized by uncontrolled cell growth and proliferation, cancer encompasses a diverse array of malignancies, each with its unique molecular and clinical characteristics [1]. Despite decades of intensive research efforts and significant advancements in diagnostic techniques and therapeutic modalities, cancer remains a leading cause of morbidity and mortality worldwide [2,3].
The complexity of cancer arises from its multifaceted nature, encompassing a spectrum of biological, genetic, and environmental factors that contribute to disease initiation, progression, and therapeutic response [4]. Tumors exhibit remarkable heterogeneity, both within individual malignancies and across different cancer types, leading to diverse clinical presentations and treatment outcomes [5]. This intrinsic variability poses significant challenges for the development of effective and universally applicable treatment strategies [6].
Conventional cancer therapies, including surgery, chemotherapy, and radiation therapy, have long been the cornerstone of cancer management [7]. While these modalities have demonstrated efficacy in certain contexts, they are associated with considerable limitations, such as systemic toxicity, treatment resistance, and adverse side effects [8].
In recent years, there has been growing recognition of the importance of exploring new paradigms in cancer therapy to address the limitations of traditional treatments. One promising avenue is the repurposing of existing drugs for anticancer purposes. Drug repurposing, also known as drug repositioning, involves identifying new therapeutic indications for drugs that have already been approved for other medical conditions [9,10]. By taking advantage of the existing pharmacological and safety profiles of these drugs, researchers can speed up the development of new anticancer therapies and potentially bypass the time-consuming and costly process of drug discovery and development [11,12]. A key advantage of drug repurposing lies in its speed of development. Unlike de novo drug discovery, which can take years or even decades to bring a new drug to market, repurposed drugs have already undergone safety and pharmacology tests in humans. This enables a more rapid transition from preclinical studies to clinical trials, accelerating the availability of new treatment options for cancer patients [13,14]. Furthermore, the wide diversity of molecules available for repurposing offers a wealth of potential candidates, each with unique mechanisms of action and safety profiles. This diversity provides researchers with a rich pool of compounds to explore, increasing the likelihood of identifying effective anticancer agents. Over the years, several drugs initially developed for other indications have been successfully repurposed to treat other diseases, demonstrating the potential of this strategy (Table 1).
In addition, there is also growing interest in exploring combination therapies as a strategy to increase treatment efficacy and overcome drug resistance in cancer [24]. Combination therapy involves the simultaneous or sequential administration of two or more drugs with complementary mechanisms of action [25]. By targeting multiple pathways involved in cancer growth and progression, combination therapy has the potential to achieve synergistic effects, maximizing therapeutic benefit while minimizing adverse effects [25,26]. Moreover, repurposed drugs can be combined with other conventional or experimental therapies, thereby broadening treatment options and enabling therapeutic synergies [27]. For example, metformin, originally used to treat type 2 diabetes, has been combined with chemotherapy and radiotherapy to improve outcomes in various cancers [28]. Similarly, sildenafil, known for treating erectile dysfunction, has been combined with chemotherapeutic agents like docetaxel to enhance drug delivery to tumors [29]. Thalidomide, once a treatment for morning sickness, has been repurposed for multiple myeloma and shows significant efficacy when combined with dexamethasone [30]. Digoxin, traditionally used for heart failure, enhances the effects of radiation therapy in hypoxic tumor environments by inhibiting HIF-1α [31,32]. This combined approach is particularly relevant for overcoming drug resistance and controlling the complex signaling pathways involved in cancer progression, offering new hope for improved outcomes in patients with advanced or refractory disease.
The importance of studies on new drugs for cancer treatment has grown significantly in recent decades due to its relevance [33]. Several works of our group have explored the cytotoxic effects of repurposed drugs and presented positive results regarding the cytotoxicity of some repurposed drugs, such as atorvastatin and nitrofurantoin in breast cancer and neuroblastoma [34] and CNS drugs in bladder cancer [35].
Although these studies have contributed significantly to the discovery of new possible drugs for the treatment of cancer, it is still important to study new possible treatments to try to reduce the therapeutic dose of chemotherapy and, consequently, its side effects.
In this context, our work proposes exploring the concept of repurposing positive inotropic agents in cancer therapy. Notably, some positive inotropic agents have already demonstrated anti-tumor activity in various studies, suggesting their potential as novel therapeutic agents in cancer treatment [31,36,37,38,39,40,41]. The main novelty of this study lies in the fact that this class of drugs has not been widely studied in the context of cancer treatment. By elucidating the opportunities inherent in this approach, we aim to contribute to the ongoing efforts to advance cancer treatment and improve patient care.

2. Positive Inotropic Agents

Positive inotropic agents are medications that exert a positive effect on cardiac contractility, meaning they increase the force of contraction of the heart muscle. These agents are commonly used in the management of heart failure and other cardiovascular conditions characterized by impaired cardiac function. By enhancing myocardial contractility, positive inotropic agents improve cardiac output and alleviate symptoms such as fatigue, dyspnea, and exercise intolerance. It is important to note that cardiovascular disease is currently the leading cause of global mortality [42].
The mechanism of action of positive inotropic agents primarily involves modulation of the intracellular calcium levels in cardiac myocytes (Figure 1). These agents typically act by increasing the intracellular concentration of calcium ions, which in turn enhances the interaction between actin and myosin filaments, leading to stronger and more efficient contraction of the heart muscle.

2.1. Cardiac Glycosides

One of the most well-known positive inotropic agents is digoxin, a cardiac glycoside derived from the foxglove plant, known as Digitalis lanata [43]. Digoxin works by inhibiting the sodium-potassium ATPase pump in cardiac myocytes, resulting in an increase in intracellular calcium levels and enhanced myocardial contractility [44]. Approved by the FDA in 1954, digoxin is used to treat different cardiac conditions like atrial flutter, atrial fibrillation, and congestive heart failure along with their related symptoms, as well as the termination of fetal life before an abortion [45,46]. Despite its long history of use, digoxin is now less commonly prescribed due to concerns about its narrow therapeutic window and potential for toxicity [47]. More advanced treatments with fewer side effects and improved safety profiles, such as beta-blockers, phosphodiesterase (PDE) inhibitors, and calcium-channel blockers, have superseded its use.

2.2. β-Agonists

β-agonists, also known as β-adrenergic agonists or β-adrenergic receptor agonists, exert their effects by stimulating beta-adrenergic receptors in the heart. These receptors, when activated, lead to an increase in myocardial contractility, heart rate, and conduction velocity. Dopamine and dobutamine are both positive inotropic agents that are commonly used in the management of various cardiovascular conditions, particularly in acute settings such as heart failure and shock states. β-adrenergic agonists bind to and activate beta-adrenergic receptors located on the cell membrane of target cells. Dopamine primarily acts on beta-1 adrenergic receptors in the heart. Upon receptor activation, beta-adrenergic agonists initiate intracellular signaling cascades through activation of the G proteins associated with the receptor. This leads to the activation of adenylate cyclase, an enzyme responsible for catalyzing the conversion of adenosine triphosphate (ATP) to cyclic adenosine monophosphate (cAMP). The production of cAMP serves as a second messenger that mediates the cellular effects of beta-adrenergic receptor activation [48]. Elevated levels of cAMP activate protein kinase A (PKA), which phosphorylates various proteins within the cell. Phosphorylation of target proteins by PKA results in a wide range of cellular responses, depending on the tissue type [49]. In cardiac myocytes, beta-1 adrenergic receptor activation leads to increased calcium influx through voltage-gated calcium channels, enhancing myocardial contractility (positive inotropic effect) [50]. In vascular smooth muscle cells, beta-adrenergic receptor activation can lead to vasodilation (particularly via beta-2 receptors), relaxation of bronchial smooth muscle (bronchodilation), and other effects [51].

2.3. Phosphodiesterase (PDE) Inhibitors

Another class of positive inotropic agents comprises phosphodiesterase (PDE) inhibitors, including milrinone, amrinone, enoximone, and cilostazol. These drugs function by impeding the degradation of cAMP and cyclic guanosine monophosphate (cGMP) within cells [52]. Elevated levels of cAMP and cGMP activate PKA and protein kinase G (PKG), respectively, by binding to the regulatory subunits of these kinases. Subsequently, PKA and PKG phosphorylate various intracellular proteins, initiating a cascade of events [53]. In cardiac myocytes, heightened cAMP levels prompt the activation of PKA, leading to increased calcium influx through L-type calcium channels and augmented calcium release from the sarcoplasmic reticulum [54]. Consequently, this enhances myocardial contractility, resulting in a positive inotropic effect. Concurrently, elevated cGMP levels activate PKG, which phosphorylates proteins involved in smooth muscle relaxation and vasodilation. This leads to the relaxation of vascular smooth muscle, promoting vasodilation and a decrease in systemic vascular resistance [55].

2.4. Calcium Sensitizers

Levosimendan is another positive inotropic agent that has gained attention for its unique mechanism of action. Unlike traditional agents that primarily increase intracellular calcium levels, levosimendan exerts its effects by sensitizing cardiac myofilaments to calcium, thereby enhancing myocardial contractility without increasing the intracellular calcium concentration [56]. Additionally, levosimendan exhibits selective inhibition of PDE, which could potentially enhance its inotropic effects under specific experimental circumstances [57]. Furthermore, investigations conducted on porcine coronary endothelial cells proposed an additional mechanism for vasodilation involving the nitric oxide (NO) and cGMP-protein kinase G-myosin light chain kinase pathway [58]. This mechanism further amplifies the vasorelaxation effects of levosimendan.

3. Preclinical Evidence of Anti-Tumor Activity and Current Clinical Landscape

3.1. Dopamine

As one of the primary neurotransmitters in mammals, dopamine (DA), chemically known as 4-(2-aminoethyl)-1,2-benzenediol, plays a crucial role in neural signaling, with a widespread impact on various physiological functions within the central nervous system (CNS) [59]. These functions extend beyond voluntary movement and encompass reward processing, sleep regulation, appetite control, emotional expression, attention, cognitive abilities, sense of smell, visual processing, hormonal balance, and regulation of the sympathetic nervous system [60]. DA concentrations in tumor tissues are typically lower compared with normal tissues, and elevating dopamine levels through treatment seems to impede the proliferation of tumor cells. Furthermore, studies by Moreno-Smith et al. have demonstrated that dopamine diminishes angiogenesis induced by chronic stress, thereby mitigating tumor expansion in ovarian carcinoma [36]. Inhibition of angiogenesis was also seen in gastric cancer with dopamine treatment by reducing tumor growth [37]. In another study, DA was shown to inhibit the vascular permeabilizing strongly and selectively and the angiogenic activities of VPF and VEGF. Via DA D2 receptors, induced the endocytosis of VEGF receptor 2, which is critical for promoting angiogenesis, thus preventing VPF/VEGF binding, receptor phosphorylation, and subsequent signaling steps [61]. Sarkar et al. presented results showing that the administration of low doses of DA, similar to the doses used in human therapies, prevents the progression of breast and colon cancers in vitro. Furthermore, a synergistic result is observed when DA is combined with conventional anticancer drugs (5-fluorouracil), resulting in prolonged survival of tumor-bearing animals [62]. Recently, a group of researchers successfully fabricated cobalt-ferrite (CF) nanoparticles (NPs) as a DA delivery agent by functionalizing CF with DA and magnetically manipulated polyethylene glycol (PEG) as an anticancer drug delivery agent for the administration of DA via a thermal and pH-sensitive form. They found that CF-DA-PEG is able to successfully transport DA inside A549 lung cancer cell lines and increases cancer cell apoptosis by inducing ROS, mitochondrial dysfunction, and destruction of the actin cytoskeleton. This study shows a new way of treating cancer by releasing DA into the cancer region at a low pH and high temperature with minimal side effects [63].

3.2. Dobutamine

Dobutamine is a β-1 agonist approved by the FDA in 1978 for short-term use in patients with decreased contractility caused by heart failure or cardiac surgical procedures, leading to cardiac decompensation [64,65]. In 2011, Bao et al. found that dobutamine inhibits the yes-associated protein (YAP)-dependent gene transcription, which has been observed in a number of types of tumors [66,67,68,69,70]. With this, Yin et al. decided to study the effect of dobutamine on the proliferation, apoptosis, and invasiveness of the MG-63 human osteosarcoma cell line. Dobutamine was able to significantly inhibit the proliferation of MG-63 osteosarcoma cells in a time- and concentration-dependent manner. In addition, it was able to increase cell apoptosis and arrest the cell cycle at the G2/M transition stage, reduce migration and invasion, and induce expression of caspases 3 and 9 [38]. Another study conducted by Xie and his team was able to prove the antitumor effect of dobutamine on multiple myeloma cells, including inhibition of cell proliferation, promotion of apoptosis, and induction of cell cycle arrest at the G1 phase in a dose-dependent manner via inhibiting activation of the MAPK signaling pathway [39]. In human gastric adenocarcinoma, dobutamine was able to significantly inhibit cell growth, migration, cell colony formation, and invasion. In addition, dobutamine interrupted the cell cycle in the G1 or S phase and increased the apoptosis rate of gastric adenocarcinoma SGC-7901 cells [71].

3.3. Digoxin

Digoxin, known for its role as a cardioprotective drug due to its inhibition of Na+/K+-ATPase, hence promoting the intracellular accumulation of Ca2+ ions, has also garnered attention for its potential anticancer properties [31,40,72,73,74,75,76]. Interest in the relationship between cardiac glycosides (CGs) and cancer began to grow in the late 1970s due to two key discoveries. Firstly, it was observed that malignant cells display changes in the activity of the Na+/K+-ATPase [77]. Secondly, certain CGs were identified as genuine phytoestrogens, suggesting a potential impact on the development and progression of hormone-sensitive cancers such as breast carcinoma [78]. In 2012, Platz et al. conducted a study for the identification of novel indication for commonly prescribed drugs. They explored the potential of clinically used drugs for the treatment of prostate cancer through a combination of efficient, high-throughput laboratory-based screening and a large prospective cohort study. Digoxin has emerged as the leading candidate due to its significant potency in inhibiting proliferation in vitro (IC50 = 163 nM) and widespread clinical use [74]. Digoxin has been shown to significantly inhibit the growth of pancreatic cancer cells [79]. Notably, it exhibits low cytotoxicity in normal cells at low concentrations [80]. Moreover, digoxin has demonstrated efficacy in inhibiting primary tumor growth and metastasis in breast cancer cells implanted into SCID mice [81]. Another study demonstrated that digoxin can inhibit proto-oncogene tyrosine-protein kinase Src and the phosphorylation and expression of epidermal growth factor receptor (EGFR) as well as signal transducer and activator of transcription 3 (STAT3). This inhibition contributes to the suppression of cancer cell proliferation, migration, and invasion [82]. In non-small cell lung cancer (NSCLC) cells, digoxin downregulates NDRG1 and VEGF by inhibiting HIF-1α under hypoxic conditions and induces autophagy, thereby inhibiting cell growth through regulation of the mTOR and ERK1/2 signaling pathways [83,84]. In small cell lung cancer (SCLC), six cell lines were subjected to digoxin treatment, all exhibiting high sensitivity with rather low IC50 values (approximately 50 nM). It was identified that digoxin treatment alone induced cell cycle arrest in the G2 or M phase and triggered apoptosis across these cell lines [76].

3.4. Cilostazol

Cilostazol, primarily utilized to treat intermittent claudication, is a derivative of 2-oxyquinolone. Its mechanism involves inhibiting phosphodiesterase III, leading to increased levels of cyclic adenosine monophosphate (cAMP). Its multifaceted pharmacological profile includes antiplatelet, antiproliferative, vasodilatory, and ischemic-reperfusion protective effects [85]. In a study conducted in 1998 by Ikeda et al., it was found that administering cilostazol significantly prolonged the survival of rats with liver metastatic tumors [86]. The researchers proposed that the interaction between cancer cells and platelets plays a crucial role in the metastatic process. They suggested that cilostazol’s ability to inhibit platelet function could account for the observed improvement in their experimental outcomes. The data presented by Murata et al. clearly indicate that cilostazol directly inhibits colon cancer cell motility. Indeed, the study revealed that cilostazol effectively hinders trans-cellular migration of DLD-1 cells through mesothelial cell monolayers induced with 10% FBS, implying potential benefits for host cells [87]. In another study, cilostazol was shown to cause anticancer effects in human squamous cell carcinoma and colon cancer cells [88]. Using Hep3B and SK-Hep1 cells, cilostazol demonstrated the ability to inhibit hepatocellular carcinoma (HCC) cell proliferation through multiple mechanisms. It induced apoptosis, arrested the cell cycle at the G0 or G1 phase, and reduced the expression of cyclin D1. The antiproliferative effects of cilostazol have been linked to the activation of AMP-activated protein kinase (AMPK) and inhibition of extracellular signal-regulated kinase (ERK) and AKT signaling pathways (Figure 2).
Liu et al. observed that cilostazol eliminates radiation-resistant glioblastoma [41]. In glioblastoma multiforme (GBM) cells, which are radiation-resistant, there was an increase in cell mobility and an acceleration in cell proliferation. Notably, GBM cells typically exhibit high expression and activation of the large conductance calcium-activated potassium channel (BK channel). However, radiation resistance resulted in a reduction in BK channel activity, regardless of its expression level. Cilostazol has shown promising results, being able to inhibit cell motility, proliferation, and the formation of tumor spheres in GBM cells’ radiation resistance. In addition, in an in vivo model of radiation-resistant GBM cells, cilostazol slowed tumor growth in vivo and prolonged survival in this model.

3.5. Levosimendan

Levosimendan is renowned for its efficacy in managing acute decompensated heart failure (ADHF) by enhancing myocardial contractility and exerting vasodilatory effects through the stimulation of cardiac troponin C and opening of ATP-sensitive potassium channels. Despite the lack of any studies on levosimendan for cancer treatment, our group initiated a study to investigate the potential benefits of combining levosimendan with 5-FU to enhance the efficacy of 5-FU and reduce its required dose in the treatment of urological cancers. Thus, levosimendan demonstrated significant anticancer effects on its own in prostate cancer (PC-3) and bladder cancer (UM-UC-5) cells. When combined with 5-FU, levosimendan potentiated the anticancer effects in both cell lines. Levosimendan can inhibit cell migration, proliferation, and colony formation, possibly affecting cyclic adenosine monophosphate (cAMP) levels and nitric oxide (NO) production. Therefore, the combination of levosimendan with 5-FU shows promising potential as a form of therapy for urological cancers, offering an innovative approach to improving patient outcomes. This combination strategy may allow for a reduction in individual drug doses and, consequently, adverse effects while maintaining therapeutic efficacy. The inhibition of cancer cell migration and colony formation suggests a possible role for levosimendan in metastasis prevention and tumor growth control.
Despite promising findings in preclinical studies investigating the anti-tumor potential of positive inotropic agents, it is important to note that specific clinical trials in this area have not been identified to date. The absence of current clinical trials underscores the need for further research initiatives to assess the therapeutic potential of positive inotropic agents in cancer treatment. These efforts may include the development of prospective randomized clinical trials investigating the effects of positive inotropic agents in patients with different types of cancer, as well as the evaluation of their safety, tolerability, and efficacy in combination with other therapeutic modalities. While preclinical studies provide a solid foundation for investigating the anti-tumor potential of positive inotropic agents, well-designed clinical trials are essential to validate these results and determine their role in cancer treatment.

4. Repurposing Positive Inotropic Agents for Oncology

Positive inotropic agents such as dopamine, dobutamine, digoxin, cilostazol, and levosimendan have demonstrated potential anticancer properties through various mechanisms. These agents not only improve cardiac function but also exhibit anti-tumor activities by influencing cellular pathways. Based on previous research elucidating the anti-tumor effects of levosimendan, this article explores the shared mechanisms (Table 2) underlying the anticancer properties of positive inotropic agents [12].

4.1. Inhibition of Phosphodiesterase 3

Phosphodiesterase 3 (PDE3) is widely distributed across various tissues and exists in two distinct forms, known as isoenzymes PDE3A and PDE3B. The activity of PDE3 isoenzymes plays a crucial role in various physiological processes, spanning cardiovascular function, insulin secretion, carbohydrate metabolism, lipolysis, and platelet aggregation. PDE3A exhibits predominant expression in several key tissues including the heart, vascular smooth muscle, platelets, pancreas, gastrointestinal (GI) tract, endometrium, fallopian tubes, oocytes, and kidney [89]. In vascular smooth muscle, PDE3A plays a crucial role in modulating contractions, contributing to the regulation of vascular tone and blood pressure. Additionally, in platelets, PDE3A activity is involved in the process of aggregation, influencing hemostasis and thrombosis [90,91]. Specifically, the catalytic functions of PDE3B isoenzymes are subject to activation through phosphorylation by PKA and the PI3K/PKB pathways in response to diverse hormonal signals. This regulatory mechanism underscores the versatility of PDE3 in modulating intracellular signaling cascades in different cellular contexts. Moreover, PDE3 exerts physiological significance in numerous tissues, including the heart, smooth muscle, platelets, liver, oocytes, and adipocytes. Its presence and activity in these tissues highlight its multifaceted roles in regulating cellular responses to various stimuli and underscores its importance in maintaining homeostasis across different organ systems [92]. Inhibition of the PDE isoenzyme 3 leads to an increase in the intracellular concentrations of the second messenger cyclic adenosine monophosphate (cAMP).

Increase in cAMP Levels

Previous studies have indeed demonstrated that increasing levels of cAMP in cancer cells can inhibit their transcellular migration in vitro [93,94,95]. Overall, drugs that target cAMP signaling pathways represent a promising approach for preventing metastasis in cancer patients.
Existing extensively in cells, cAMP serves as a critical intracellular second messenger in various cellular processes, influencing cell metabolism, ion channel activity, gene expression, cell growth, differentiation, and apoptosis [96]. The major targets of cAMP include PKA, exchange proteins activated by cAMP (Epac1 and Epac2), and ion channels. PKA plays a crucial role in the regulation of cell proliferation by exerting its effects on transcription factors and other signaling molecules. One mechanism by which PKA may inhibit proliferation is interfering with the Ras-Raf-MEK-ERK signaling pathway, which is a key regulator of cell growth and proliferation. Specifically, PKA can phosphorylate and thereby inhibit the activity of c-Raf, a downstream effector of Ras. By phosphorylating c-Raf, PKA can prevent its activation and subsequent downstream signaling events, including the activation of MEK and ERK kinases. This inhibition of the Ras-Raf-MEK-ERK pathway by PKA can lead to decreased proliferation and cell growth [97,98]. Epac proteins, as a major effectors of cAMP signaling, participate in a wide array of cellular functions mediated by cAMP, such as cell adhesion [99,100], the cell-cell junction [101,102], cell differentiation [103], cell proliferation [104] and apoptosis [105]. Ion channels that are directly regulated by cAMP are permeable to cations such as sodium (Na+) and calcium (Ca2+), and their activation by cAMP leads to changes in membrane potential and cellular excitability. An increase in intracellular sodium concentration has been reported early in the cell death process, particularly in the context of apoptosis [106,107,108,109].

4.2. Production of Nitric Oxide

Nitric oxide (NO) is a ubiquitous free radical signaling molecule that plays a fundamental role in regulating numerous cellular processes. Its diverse functions include the modulation of angiogenesis, smooth muscle tone, immune response, apoptosis, and synaptic communication [110]. NO production in cancer is a multifaceted process with both pro-tumorigenic and anti-tumorigenic effects, depending on various factors such as the stage of cancer, the tumor microenvironment, and the concentration of NO (Figure 3). At low concentrations, NO functions mainly as a signaling molecule through the nitrosylation of soluble guanylate cyclase (sGC), leading to an increase in cyclic guanosine monophosphate (cGMP) levels. This activation of sGC results in vasodilation and may also promote angiogenesis through the activation of signaling pathways involving hypoxia-induced factor 1 alpha (HIF1α) and vascular endothelial growth factor (VEGF) [111]. These effects can promote tumor growth and metastasis. However, at higher concentrations, NO has been shown to inhibit cell proliferation and induce apoptosis, ultimately leading to the death of cancer cells [112,113].
NO can induce the expression of the tumor suppressor gene p53, which is a critical regulator of the cell cycle and apoptosis [114]. Meanwhile, p53 activation leads to the transcription of several pro-apoptotic genes, including Bax [115]. The pro-apoptotic protein Bax promotes the activation of caspases such as caspase 3, a critical executioner caspase that is activated downstream of mitochondrial events such as the release of cytochrome c, leading to apoptotic cell death [116]. NO-induced activation of caspase 8 can cleave Bid into tBid, which then translocates to mitochondria to promote cytochrome c release and further caspase activation [117].

4.3. Reduction in Reactive Oxygen Species Levels

Reactive oxygen species (ROS) are a group of reactive molecules that contain oxygen and are considered to be natural byproducts of cellular metabolism primarily produced in the mitochondria [118,119]. They include molecules such as superoxide anion (O2·−), hydroxyl radical (·OH), and hydrogen peroxide (H2O2) [120]. While ROS act as signaling molecules in various cellular pathways, influencing processes such as cell proliferation, differentiation, apoptosis, and inflammation and playing a role in host defense mechanisms, where excessive accumulation can lead to oxidative stress, causing damage to proteins, lipids, and DNA [121,122,123]. This imbalance between ROS production and antioxidant defense mechanisms can result from various factors, including environmental stressors (such as UV and ionizing radiation), metabolic dysregulation, and inflammation [124]. Oxidative stress contributes to cellular damage by oxidizing biomolecules, disrupting cellular function [125], and promoting the development of various diseases, including cancer [126,127,128,129,130,131,132], neurodegenerative disorders [133,134], and cardiovascular diseases [135,136]. When cancer cells undergo various cellular processes, their levels of ROS tend to increase, resulting in oxidative stress. This heightened oxidative stress triggers the activation of specific redox-sensitive transcription factors and growth regulatory proteins, including NF-kB, AP-1, HIF-1A, and NRF2. Activation of these transcription factors disrupts signaling pathways and alters the expression of oxidative and inflammatory mediators, which are crucial in cancer development and treatment complexities [137,138]. Moreover, cancer cells release various mediators, including oxidative and inflammatory components, which orchestrate changes in surrounding cells and tissues within the tumor microenvironment (TME). The TME, comprising diverse immune cells (both innate and adaptive) and fibroblasts, emerged as a critical determinant in cancer progression, metastasis, and clinical outcomes [139]. ROS play a pivotal role in driving signaling pathways associated with epithelial-mesenchymal transition (EMT), metastasis, and adverse clinical consequences [140,141]. Additionally, metabolic reprogramming is a key aspect of cancer pathogenesis, ensuring a continuous energy supply for uncontrolled growth, proliferation, and maintenance of stem cell-like properties in cancer cells. Extensive research has highlighted the significant contribution of ROS to reshaping metabolic dynamics across various cancer types, including lymphoid and myeloid cells [142,143,144,145]. In recent years, numerous clinical trials have investigated therapeutic drugs aimed at regulating ROS in cancer cells [146]. Additionally, there has been an extensive review of molecules that manage ROS homeostasis for cancer treatment [147,148]. Several FDA-approved drugs that target ROS regulation have been repurposed for cancer treatment, presenting a novel and effective approach to cancer therapy [149]. The production of intracellular ROS was markedly lowered in human microglial HMC3 cells treated with either levosimendan or dobutamine compared with cells that were not treated. This suggests that these drugs might be effective in reducing ROS buildup [150]. In another study conducted by Zhao et al., dimethyl succinate was used to trigger total ROS production, which was significantly inhibited when treated with digoxin [151].
Reducing ROS levels in cancer cells involves multiple mechanisms, each contributing to improved therapeutic outcomes. Antioxidant enzymes such as superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), and peroxiredoxins (Prxs) play a crucial role in mitigating oxidative stress by converting harmful ROS into less reactive molecules [152]. For instance, SOD converts superoxide radicals into hydrogen peroxide, which catalase then breaks down into water and oxygen, while GPx reduces hydrogen peroxide and lipid hydroperoxides using glutathione, and Prx reduces peroxynitrite and organic hydroperoxides [153]. Reducing ROS levels sensitizes cancer cells to conventional therapies such as chemotherapy and radiotherapy. Reducing ROS alters the cellular environment, making cancer cells more susceptible to the cytotoxic effects of these treatments [154]. In addition, the tumor microenvironment undergoes significant changes when ROS levels are reduced. Lower levels of oxidative stress create a more favorable environment for the immune system to function [125]. Cancer cells often exploit ROS to evade immune surveillance and mount immunosuppressive responses. By reducing ROS, the immune system gains the upper hand, mounting more effective anti-tumor responses [155].

5. Challenges and Future Opportunities

The use of positive inotropic agents in cancer therapy presents a unique set of challenges and opportunities that demand a careful and innovative approach. One of the main challenges lies in identifying and developing effective therapeutic agents that can tackle the dual threat of heart disease, the leading cause of death worldwide, and cancer, one of the most lethal diseases, not only improving cardiac function but also displaying robust anti-tumor properties. The complexity of tumor biology and the heterogeneity of tumors require an in-depth understanding of the mechanisms by which these agents exert their anti-tumor effects, as well as their potential toxicity and bioavailability.
Understanding the anti-tumor mechanisms of these agents is crucial, as it could reveal new therapeutic targets and treatment strategies. However, elucidating these mechanisms is a significant challenge due to their multifunctionality and complex interaction with cell signaling pathways. In addition, identifying predictive biomarkers that can help select the patients most likely to respond to treatment with positive inotropic agents is essential to maximize therapeutic benefits and minimize adverse effects. The tables below (Table 3 and Table 4) provide a comprehensive comparison of the featured inotropic agents (dopamine, dobutamine, digoxin, cilostazol, and levosimendan) and standard chemotherapy drugs used in cancer treatment.
Drug repurposing strategies can yield significant advantages in terms of overall development cost, time to market, and mitigating failure rates attributed to safety concerns. In certain instances, repurposed indications are uncovered either by the original drug developer during pre-clinical investigations or incidentally observed as a side effect in clinical trials, subsequently leading to patenting. A notable illustration of this phenomenon occurred during development of the hypertensive medication sildenafil, which was redirected toward the creation of Viagra for treating erectile dysfunction following the discovery of potent off-target effects in clinical trials [156].
The safety and tolerability of positive inotropic agents in cancer patients are paramount considerations. Given that these agents are FDA-approved and have been in use for many years, they are generally regarded as safe. However, it is crucial to acknowledge that cancer patients often experience a significant burden of toxicity from chemotherapy and radiotherapy. Therefore, careful assessment of potential side effects and effective symptom management remain essential. While positive inotropic agents are well-established medications, ongoing research efforts are still needed to optimize strategies for mitigating adverse effects and improving patients’ quality of life.
Moreover, the combination therapy of positive inotropic agents with other treatment modalities, such as chemotherapy, immunotherapy, and target therapy, offers a promising opportunity to improve clinical outcomes in cancer patients. Identifying synergistic therapeutic combinations and appropriate sequencing strategies can increase treatment efficacy and consequently reduce the required dose of chemotherapy, for example, and reduce the development of resistance.

6. Conclusions

The exploration of positive inotropic agents as potential candidates for cancer therapy represents a compelling avenue for innovation in the field of oncology. Despite the well-established use of these agents in cardiovascular medicine, their therapeutic potential in cancer treatment is an emerging area of research with promising preclinical evidence.
The multifaceted mechanisms of action exhibited by positive inotropic agents, including their ability to modulate intracellular signaling pathways, increase cAMP levels, produce nitric oxide, and decrease reactive oxygen species, highlight their potential utility as anticancer agents. Furthermore, their established safety profiles and FDA approval for other indications offer a valuable foundation for further investigation in oncology.
However, several challenges must be addressed to fully realize the therapeutic potential of positive inotropic agents in cancer treatment. These include elucidating the specific mechanisms underlying their anti-tumor effects, identifying predictive biomarkers for patient selection, and optimizing strategies to mitigate potential adverse effects in cancer patients, particularly those already burdened by chemotherapy and radiotherapy toxicity. Despite these challenges, the prospect of combining positive inotropic agents with existing cancer therapies, such as chemotherapy, immunotherapy, and targeted therapy, holds great promise for improving clinical outcomes and enhancing patient care. By leveraging the synergistic effects of combination therapy, it may be possible to maximize treatment efficacy while minimizing adverse effects and drug resistance.

Author Contributions

Conceptualization, N.V.; methodology E.R.; formal analysis, E.R. and N.V.; investigation, E.R. and N.V.; writing—original draft preparation, E.R.; writing—review and editing, N.V.; supervision, N.V.; project administration, N.V.; funding acquisition, N.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financed by Fundo Europeu de Desenvolvimento Regional (FEDER) funds through the COMPETE 2020 Operational Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by the Portuguese fund CHAIR in Onco-Innovation from the Faculty of Medicine of the University of Porto (FMUP).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

E.R. acknowledges CHAIR in Onco-Innovation and the FMUP for funding their PhD grant.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brown, J.S.; Amend, S.R.; Austin, R.H.; Gatenby, R.A.; Hammarlund, E.U.; Pienta, K.J. Updating the Definition of Cancer. Mol. Cancer Res. 2023, 21, 1142–1147. [Google Scholar] [CrossRef] [PubMed]
  2. Pulumati, A.; Pulumati, A.; Dwarakanath, B.S.; Verma, A.; Papineni, R.V.L. Technological advancements in cancer diagnostics: Improvements and limitations. Cancer Rep. 2023, 6, e1764. [Google Scholar] [CrossRef] [PubMed]
  3. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.W. Cancer and radiation therapy: Current advances and future directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef] [PubMed]
  4. Derbal, Y. The Adaptive Complexity of Cancer. BioMed Res. Int. 2018, 2018, 5837235. [Google Scholar] [CrossRef] [PubMed]
  5. Gerdes, M.J.; Sood, A.; Sevinsky, C.; Pris, A.D.; Zavodszky, M.I.; Ginty, F. Emerging understanding of multiscale tumor heterogeneity. Front. Oncol. 2014, 4, 366. [Google Scholar] [CrossRef] [PubMed]
  6. Bhatia, S.; Frangioni, J.V.; Hoffman, R.M.; Iafrate, A.J.; Polyak, K. The challenges posed by cancer heterogeneity. Nat. Biotechnol. 2012, 30, 604–610. [Google Scholar] [CrossRef] [PubMed]
  7. Charmsaz, S.; Collins, D.M.; Perry, A.S.; Prencipe, M. Novel Strategies for Cancer Treatment: Highlights from the 55th IACR Annual Conference. Cancers 2019, 11, 1125. [Google Scholar] [CrossRef] [PubMed]
  8. Ahire, V.; Ahmadi Bidakhvidi, N.; Boterberg, T.; Chaudhary, P.; Chevalier, F.; Daems, N.; Delbart, W.; Baatout, S.; Deroose, C.M.; Fernandez-Palomo, C.; et al. Radiobiology of Combining Radiotherapy with Other Cancer Treatment Modalities. In Radiobiology Textbook; Baatout, S., Ed.; Springer International Publishing: Cham, Switzerland, 2023; pp. 311–386. [Google Scholar]
  9. Pushpakom, S.; Iorio, F.; Eyers, P.A.; Escott, K.J.; Hopper, S.; Wells, A.; Doig, A.; Guilliams, T.; Latimer, J.; McNamee, C.; et al. Drug repurposing: Progress, challenges and recommendations. Nat. Rev. Drug Discov. 2019, 18, 41–58. [Google Scholar] [CrossRef] [PubMed]
  10. Ribeiro, E.; Vale, N. Repurposing of the Drug Tezosentan for Cancer Therapy. Curr. Issues Mol. Biol. 2023, 45, 5118–5131. [Google Scholar] [CrossRef]
  11. Okuyama, R. Advancements in Drug Repurposing: Examples in Psychiatric Medications. Int. J. Mol. Sci. 2023, 24, 11000. [Google Scholar] [CrossRef]
  12. Ribeiro, E.; Vale, N. Understanding the Clinical Use of Levosimendan and Perspectives on its Future in Oncology. Biomolecules 2023, 13, 1296. [Google Scholar] [CrossRef]
  13. Khataniar, A.; Pathak, U.; Rajkhowa, S.; Jha, A.N. A Comprehensive Review of Drug Repurposing Strategies against Known Drug Targets of COVID-19. COVID 2022, 2, 148–167. [Google Scholar] [CrossRef]
  14. Zhang, Z.; Zhou, L.; Xie, N.; Nice, E.C.; Zhang, T.; Cui, Y.; Huang, C. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct. Target. Ther. 2020, 5, 113. [Google Scholar] [CrossRef]
  15. de Mello, T.P.; Silva, L.N.; de Souza, R.; Frota, H.F.; Branquinha, M.H.; dos Santos, A.L.S. Drug repurposing strategy against fungal biofilms. Curr. Top. Med. Chem. 2020, 20, 509–516. [Google Scholar] [CrossRef]
  16. Hernandez, J.J.; Pryszlak, M.; Smith, L.; Yanchus, C.; Kurji, N.; Shahani, V.M.; Molinski, S.V. Giving Drugs a Second Chance: Overcoming Regulatory and Financial Hurdles in Repurposing Approved Drugs As Cancer Therapeutics. Front. Oncol. 2017, 7, 273. [Google Scholar] [CrossRef]
  17. Schein, C.H. Repurposing approved drugs on the pathway to novel therapies. Med. Res. Rev. 2020, 40, 586–605. [Google Scholar] [CrossRef]
  18. Suchonwanit, P.; Thammarucha, S.; Leerunyakul, K. Minoxidil and its use in hair disorders: A review. Drug Des. Devel Ther. 2019, 13, 2777–2786. [Google Scholar] [CrossRef]
  19. Drew, D.A.; Cao, Y.; Chan, A.T. Aspirin and colorectal cancer: The promise of precision chemoprevention. Nat. Rev. Cancer 2016, 16, 173–186. [Google Scholar] [CrossRef]
  20. Latif, T.; Chauhan, N.; Khan, R.; Moran, A.; Usmani, S.Z. Thalidomide and its analogues in the treatment of Multiple Myeloma. Exp. Hematol. Oncol. 2012, 1, 27. [Google Scholar] [CrossRef]
  21. Sperling, R. Zidovudine. Infect. Dis. Obstet. Gynecol. 1998, 6, 197–203. [Google Scholar] [CrossRef]
  22. Mok, C.C. Rituximab for the treatment of rheumatoid arthritis: An update. Drug Des. Devel Ther. 2013, 8, 87–100. [Google Scholar] [CrossRef]
  23. Cummings, S.R.; Eckert, S.; Krueger, K.A.; Grady, D.; Powles, T.J.; Cauley, J.A.; Norton, L.; Nickelsen, T.; Bjarnason, N.H.; Morrow, M.; et al. The effect of raloxifene on risk of breast cancer in postmenopausal women: Results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation. JAMA 1999, 281, 2189–2197. [Google Scholar] [CrossRef]
  24. Victoir, B.; Croix, C.; Gouilleux, F.; Prié, G. Targeted Therapeutic Strategies for the Treatment of Cancer. Cancers 2024, 16, 461. [Google Scholar] [CrossRef]
  25. Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef]
  26. Delou, J.M.A.; Souza, A.S.O.; Souza, L.C.M.; Borges, H.L. Highlights in Resistance Mechanism Pathways for Combination Therapy. Cells 2019, 8, 1013. [Google Scholar] [CrossRef]
  27. Weth, F.R.; Hoggarth, G.B.; Weth, A.F.; Paterson, E.; White, M.P.J.; Tan, S.T.; Peng, L.; Gray, C. Unlocking hidden potential: Advancements, approaches, and obstacles in repurposing drugs for cancer therapy. Br. J. Cancer 2024, 130, 703–715. [Google Scholar] [CrossRef]
  28. Zhu, L.; Yang, K.; Ren, Z.; Yin, D.; Zhou, Y. Metformin as anticancer agent and adjuvant in cancer combination therapy: Current progress and future prospect. Transl. Oncol. 2024, 44, 101945. [Google Scholar] [CrossRef]
  29. Muniyan, S.; Rachagani, S.; Parte, S.; Halder, S.; Seshacharyulu, P.; Kshirsagar, P.; Siddiqui, J.A.; Vengoji, R.; Rauth, S.; Islam, R.; et al. Sildenafil Potentiates the Therapeutic Efficacy of Docetaxel in Advanced Prostate Cancer by Stimulating NO-cGMP Signaling. Clin. Cancer Res. 2020, 26, 5720–5734. [Google Scholar] [CrossRef]
  30. von Lilienfeld-Toal, M.; Hahn-Ast, C.; Furkert, K.; Hoffmann, F.; Naumann, R.; Bargou, R.; Cook, G.; Glasmacher, A. A systematic review of phase II trials of thalidomide/dexamethasone combination therapy in patients with relapsed or refractory multiple myeloma. Eur. J. Haematol. 2008, 81, 247–252. [Google Scholar] [CrossRef]
  31. Wang, Y.; Ma, Q.; Zhang, S.; Liu, H.; Zhao, B.; Du, B.; Wang, W.; Lin, P.; Zhang, Z.; Zhong, Y.; et al. Digoxin Enhances the Anticancer Effect on Non-Small Cell Lung Cancer While Reducing the Cardiotoxicity of Adriamycin. Front. Pharmacol. 2020, 11, 186. [Google Scholar] [CrossRef]
  32. Zhang, H.; Qian, D.Z.; Tan, Y.S.; Lee, K.; Gao, P.; Ren, Y.R.; Rey, S.; Hammers, H.; Chang, D.; Pili, R.; et al. Digoxin and other cardiac glycosides inhibit HIF-1alpha synthesis and block tumor growth. Proc. Natl. Acad. Sci. USA 2008, 105, 19579–19586. [Google Scholar] [CrossRef] [PubMed]
  33. Falzone, L.; Salomone, S.; Libra, M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front. Pharmacol. 2018, 9, 1300. [Google Scholar] [CrossRef]
  34. Moura, C.; Correia, A.S.; Pereira, M.; Ribeiro, E.; Santos, J.; Vale, N. Atorvastatin and Nitrofurantoin Repurposed in the Context of Breast Cancer and Neuroblastoma Cells. Biomedicines 2023, 11, 903. [Google Scholar] [CrossRef] [PubMed]
  35. Gouveia, M.J.; Ribeiro, E.; Vale, N. A Surprising Repurposing of Central Nervous System Drugs against Squamous Cell Carcinoma of the Bladder, UM-UC-5. Pharmaceutics 2024, 16, 212. [Google Scholar] [CrossRef] [PubMed]
  36. Moreno-Smith, M.; Lu, C.; Shahzad, M.M.; Pena, G.N.; Allen, J.K.; Stone, R.L.; Mangala, L.S.; Han, H.D.; Kim, H.S.; Farley, D.; et al. Dopamine blocks stress-mediated ovarian carcinoma growth. Clin. Cancer Res. 2011, 17, 3649–3659. [Google Scholar] [CrossRef] [PubMed]
  37. Chakroborty, D.; Sarkar, C.; Mitra, R.B.; Banerjee, S.; Dasgupta, P.S.; Basu, S. Depleted dopamine in gastric cancer tissues: Dopamine treatment retards growth of gastric cancer by inhibiting angiogenesis. Clin. Cancer Res. 2004, 10, 4349–4356. [Google Scholar] [CrossRef] [PubMed]
  38. Yin, J.; Dong, Q.; Zheng, M.; Xu, X.; Zou, G.; Ma, G.; Li, K. Antitumor activity of dobutamine on human osteosarcoma cells. Oncol. Lett. 2016, 11, 3676–3680. [Google Scholar] [CrossRef] [PubMed]
  39. Xie, B.; Xu, Z.; Yang, G.; Chen, G.; Li, B.; Hu, L.; Xiao, W.; Sun, X.; Gao, M.; Gao, L.; et al. Antitumor effect of dobutamine on multiple myeloma via mitogen-activated protein kinase pathway in vitro. Acta Biochim. Et Biophys. Sin. 2016, 48, 1135–1137. [Google Scholar] [CrossRef] [PubMed]
  40. Menger, L.; Vacchelli, E.; Kepp, O.; Eggermont, A.; Tartour, E.; Zitvogel, L.; Kroemer, G.; Galluzzi, L. Trial watch: Cardiac glycosides and cancer therapy. Oncoimmunology 2013, 2, e23082. [Google Scholar] [CrossRef] [PubMed]
  41. Liu, C.C.; Wu, C.L.; Yeh, I.C.; Wu, S.N.; Sze, C.I.; Gean, P.W. Cilostazol eliminates radiation-resistant glioblastoma by re-evoking big conductance calcium-activated potassium channel activity. Am. J. Cancer Res. 2021, 11, 1148–1169. [Google Scholar]
  42. Di Cesare, M.; Perel, P.; Taylor, S.; Kabudula, C.; Bixby, H.; Gaziano, T.A.; McGhie, D.V.; Mwangi, J.; Pervan, B.; Narula, J.; et al. The Heart of the World. Glob. Heart 2024, 19, 11. [Google Scholar] [CrossRef] [PubMed]
  43. Wawer, I. Chapter 4—Solid-State Measurements of Drugs and Drug Formulations. In NMR Spectroscopy in Pharmaceutical Analysis; Holzgrabe, U., Wawer, I., Diehl, B., Eds.; Elsevier: Amsterdam, The Netherlands, 2008; pp. 201–231. [Google Scholar]
  44. Gillespie, N.D.; Witham, M.D.; Struthers, A.D. CHAPTER 40—Chronic Cardiac Failure. In Brocklehurst’s Textbook of Geriatric Medicine and Gerontology, 7th ed.; Fillit, H.M., Rockwood, K., Woodhouse, K., Eds.; W.B. Saunders: Philadelphia, PA, USA, 2010; pp. 272–285. [Google Scholar]
  45. Sethi, N.J.; Nielsen, E.E.; Safi, S.; Feinberg, J.; Gluud, C.; Jakobsen, J.C. Digoxin for atrial fibrillation and atrial flutter: A systematic review with meta-analysis and trial sequential analysis of randomised clinical trials. PLoS ONE 2018, 13, e0193924. [Google Scholar] [CrossRef]
  46. Gona, S.R.; Rosenberg, J.; Fyffe-Freil, R.C.; Kozakiewicz, J.M.; Money, M.E. Review: Failure of current digoxin monitoring for toxicity: New monitoring recommendations to maintain therapeutic levels for efficacy. Front. Cardiovasc. Med. 2023, 10, 1179892. [Google Scholar] [CrossRef]
  47. Vivo, R.P.; Krim, S.R.; Perez, J.; Inklab, M.; Tenner, T., Jr.; Hodgson, J. Digoxin: Current use and approach to toxicity. Am. J. Med. Sci. 2008, 336, 423–428. [Google Scholar] [CrossRef] [PubMed]
  48. McCoshen, J.A.; Fernandes, P.A.; Boroditsky, M.L.; Allardice, J.G. Determinants of Reproductive Mortality and Preterm Childbirth. In Advances in Organ Biology; Bittar, E.E., Zakar, T., Eds.; Elsevier: Amsterdam, The Netherlands, 1996; Volume 1, pp. 195–223. [Google Scholar]
  49. Liu, Y.; Chen, J.; Fontes, S.K.; Bautista, E.N.; Cheng, Z. Physiological and pathological roles of protein kinase A in the heart. Cardiovasc. Res. 2022, 118, 386–398. [Google Scholar] [CrossRef]
  50. Wang, W.; Zhang, H.; Gao, H.; Kubo, H.; Berretta, R.M.; Chen, X.; Houser, S.R. {beta}1-Adrenergic receptor activation induces mouse cardiac myocyte death through both L-type calcium channel-dependent and -independent pathways. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H322–H331. [Google Scholar] [CrossRef] [PubMed]
  51. Barnes, P.J. Beta-adrenoceptors on smooth muscle, nerves and inflammatory cells. Life Sci. 1993, 52, 2101–2109. [Google Scholar] [CrossRef]
  52. Dimitriadis, F.; Skouros, S.; Takenaka, A.; Sofikitis, N. Chapter 15—Beneficial or Detrimental Effects of Phosphodiesterase-5 (PDE5) Inhibitors on Semen Quality and Testicular Function? In Bioenvironmental Issues Affecting Men’s Reproductive and Sexual Health; Sikka, S.C., Hellstrom, W.J.G., Eds.; Academic Press: Boston, MA, USA, 2018; pp. 243–260. [Google Scholar]
  53. Francis, S.H.; Busch, J.L.; Corbin, J.D.; Sibley, D. cGMP-dependent protein kinases and cGMP phosphodiesterases in nitric oxide and cGMP action. Pharmacol. Rev. 2010, 62, 525–563. [Google Scholar] [CrossRef]
  54. Reiken, S.; Lacampagne, A.; Zhou, H.; Kherani, A.; Lehnart, S.E.; Ward, C.; Huang, F.; Gaburjakova, M.; Gaburjakova, J.; Rosemblit, N.; et al. PKA phosphorylation activates the calcium release channel (ryanodine receptor) in skeletal muscle: Defective regulation in heart failure. J. Cell Biol. 2003, 160, 919–928. [Google Scholar] [CrossRef]
  55. Feil, R.; Lohmann, S.M.; de Jonge, H.; Walter, U.; Hofmann, F. Cyclic GMP-dependent protein kinases and the cardiovascular system: Insights from genetically modified mice. Circ. Res. 2003, 93, 907–916. [Google Scholar] [CrossRef]
  56. Levy, J.H.; Ghadimi, K.; Bailey, J.M.; Ramsay, J.G. Chapter 30—Postoperative Cardiovascular Management. In Kaplan’s Essentials of Cardiac Anesthesia, 2nd ed.; Kaplan, J.A., Ed.; Elsevier: Philadelphia, PA, USA, 2018; pp. 758–785. [Google Scholar]
  57. Orstavik, O.; Ata, S.H.; Riise, J.; Dahl, C.P.; Andersen, G.; Levy, F.O.; Skomedal, T.; Osnes, J.B.; Qvigstad, E. Inhibition of phosphodiesterase-3 by levosimendan is sufficient to account for its inotropic effect in failing human heart. Br. J. Pharmacol. 2014, 171, 5169–5181. [Google Scholar] [CrossRef] [PubMed]
  58. Grossini, E.; Molinari, C.; Caimmi, P.P.; Uberti, F.; Vacca, G. Levosimendan induces NO production through p38 MAPK, ERK and Akt in porcine coronary endothelial cells: Role for mitochondrial K(ATP) channel. Br. J. Pharmacol. 2009, 156, 250–261. [Google Scholar] [CrossRef] [PubMed]
  59. Franco, R.; Reyes-Resina, I.; Navarro, G. Dopamine in Health and Disease: Much More Than a Neurotransmitter. Biomedicines 2021, 9, 109. [Google Scholar] [CrossRef] [PubMed]
  60. Beaulieu, J.M.; Espinoza, S.; Gainetdinov, R.R. Dopamine receptors—IUPHAR Review 13. Br. J. Pharmacol. 2015, 172, 1–23. [Google Scholar] [CrossRef] [PubMed]
  61. Basu, S.; Nagy, J.A.; Pal, S.; Vasile, E.; Eckelhoefer, I.A.; Bliss, V.S.; Manseau, E.J.; Dasgupta, P.S.; Dvorak, H.F.; Mukhopadhyay, D. The neurotransmitter dopamine inhibits angiogenesis induced by vascular permeability factor/vascular endothelial growth factor. Nat. Med. 2001, 7, 569–574. [Google Scholar] [CrossRef]
  62. Sarkar, C.; Chakroborty, D.; Chowdhury, U.R.; Dasgupta, P.S.; Basu, S. Dopamine increases the efficacy of anticancer drugs in breast and colon cancer preclinical models. Clin. Cancer Res. 2008, 14, 2502–2510. [Google Scholar] [CrossRef] [PubMed]
  63. De, D.; Upadhyay, P.; Das, A.; Ghosh, A.; Adhikary, A.; Goswami, M.M. Studies on cancer cell death through delivery of dopamine as anti-cancer drug by a newly functionalized cobalt ferrite nano-carrier. Colloids Surf. A Physicochem. Eng. Asp. 2021, 627, 127202. [Google Scholar] [CrossRef]
  64. Nguyen, L.; Roth, D.M.; Shanewise, J.S.; Kaplan, J.A. Chapter 28—Discontinuing Cardiopulmonary Bypass. In Kaplan’s Essentials of Cardiac Anesthesia, 2nd ed.; Kaplan, J.A., Ed.; Elsevier: Philadelphia, PA, USA, 2018; pp. 715–740. [Google Scholar]
  65. Ruffolo, R.R. Review: The Pharmacology of Dobutamine. Am. J. Med. Sci. 1987, 294, 244–248. [Google Scholar] [CrossRef] [PubMed]
  66. Bao, Y.; Nakagawa, K.; Yang, Z.; Ikeda, M.; Withanage, K.; Ishigami-Yuasa, M.; Okuno, Y.; Hata, S.; Nishina, H.; Hata, Y. A cell-based assay to screen stimulators of the Hippo pathway reveals the inhibitory effect of dobutamine on the YAP-dependent gene transcription. J. Biochem. 2011, 150, 199–208. [Google Scholar] [CrossRef]
  67. Zender, L.; Spector, M.S.; Xue, W.; Flemming, P.; Cordon-Cardo, C.; Silke, J.; Fan, S.T.; Luk, J.M.; Wigler, M.; Hannon, G.J.; et al. Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell 2006, 125, 1253–1267. [Google Scholar] [CrossRef]
  68. Overholtzer, M.; Zhang, J.; Smolen, G.A.; Muir, B.; Li, W.; Sgroi, D.C.; Deng, C.X.; Brugge, J.S.; Haber, D.A. Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc. Natl. Acad. Sci. USA 2006, 103, 12405–12410. [Google Scholar] [CrossRef] [PubMed]
  69. Roden, R.; Wu, T.C. How will HPV vaccines affect cervical cancer? Nat. Rev. Cancer 2006, 6, 753–763. [Google Scholar] [CrossRef] [PubMed]
  70. Castle, P.E.; Dockter, J.; Giachetti, C.; Garcia, F.A.; McCormick, M.K.; Mitchell, A.L.; Holladay, E.B.; Kolk, D.P. A cross-sectional study of a prototype carcinogenic human papillomavirus E6/E7 messenger RNA assay for detection of cervical precancer and cancer. Clin. Cancer Res. 2007, 13, 2599–2605. [Google Scholar] [CrossRef] [PubMed]
  71. Zheng, H.X.; Wu, L.N.; Xiao, H.; Du, Q.; Liang, J.F. Inhibitory effects of dobutamine on human gastric adenocarcinoma. World J. Gastroenterol. 2014, 20, 17092–17099. [Google Scholar] [CrossRef] [PubMed]
  72. Frankel, A.E.; Eskiocak, U.; Gill, J.G.; Yuan, S.; Ramesh, V.; Froehlich, T.W.; Ahn, C.; Morrison, S.J. Digoxin Plus Trametinib Therapy Achieves Disease Control in BRAF Wild-Type Metastatic Melanoma Patients. Neoplasia 2017, 19, 255–260. [Google Scholar] [CrossRef] [PubMed]
  73. Huang, L.; Garrett Injac, S.; Cui, K.; Braun, F.; Lin, Q.; Du, Y.; Zhang, H.; Kogiso, M.; Lindsay, H.; Zhao, S.; et al. Systems biology-based drug repositioning identifies digoxin as a potential therapy for groups 3 and 4 medulloblastoma. Sci. Transl. Med. 2018, 10, eaat0150. [Google Scholar] [CrossRef]
  74. Platz, E.A.; Yegnasubramanian, S.; Liu, J.O.; Chong, C.R.; Shim, J.S.; Kenfield, S.A.; Stampfer, M.J.; Willett, W.C.; Giovannucci, E.; Nelson, W.G. A novel two-stage, transdisciplinary study identifies digoxin as a possible drug for prostate cancer treatment. Cancer Discov. 2011, 1, 68–77. [Google Scholar] [CrossRef] [PubMed]
  75. Yun, H.H.; Kim, S.; Kuh, H.-J.; Lee, J.-H. Downregulation of BIS sensitizes A549 cells for digoxin-mediated inhibition of invasion and migration by the STAT3-dependent pathway. Biochem. Biophys. Res. Commun. 2020, 524, 643–648. [Google Scholar] [CrossRef] [PubMed]
  76. Deng, K.; Shen, J.; Wang, W.; Li, M.; Li, H.; Chen, C.; Zhao, H.; Zhang, M.; Xue, T.; Liu, Q.; et al. Sodium chloride (NaCl) potentiates digoxin-induced anti-tumor activity in small cell lung cancer. Cancer Biol. Ther. 2019, 20, 52–64. [Google Scholar] [CrossRef]
  77. Shen, S.S.; Hamamoto, S.T.; Bern, H.A.; Steinhardt, R.A. Alteration of sodium transport in mouse mammary epithelium associated with neoplastic transformation. Cancer Res. 1978, 38, 1356–1361. [Google Scholar]
  78. Cove, D.H.; Barker, G.A. Digoxin and hormone receptors. Lancet 1979, 2, 204. [Google Scholar] [CrossRef]
  79. Prassas, I.; Karagiannis, G.S.; Batruch, I.; Dimitromanolakis, A.; Datti, A.; Diamandis, E.P. Digitoxin-induced cytotoxicity in cancer cells is mediated through distinct kinase and interferon signaling networks. Mol. Cancer Ther. 2011, 10, 2083–2093. [Google Scholar] [CrossRef]
  80. Prassas, I.; Diamandis, E.P. Novel therapeutic applications of cardiac glycosides. Nat. Rev. Drug Discov. 2008, 7, 926–935. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, J.; Jiang, Y.; Chen, L.; Qian, Z.; Zhang, Y. Associations between HIFs and tumor immune checkpoints: Mechanism and therapy. Discov. Oncol. 2024, 15, 2. [Google Scholar] [CrossRef]
  82. Lin, S.Y.; Chang, H.H.; Lai, Y.H.; Lin, C.H.; Chen, M.H.; Chang, G.C.; Tsai, M.F.; Chen, J.J. Digoxin Suppresses Tumor Malignancy through Inhibiting Multiple Src-Related Signaling Pathways in Non-Small Cell Lung Cancer. PLoS ONE 2015, 10, e0123305. [Google Scholar] [CrossRef] [PubMed]
  83. Wei, D.; Peng, J.J.; Gao, H.; Li, H.; Li, D.; Tan, Y.; Zhang, T. Digoxin downregulates NDRG1 and VEGF through the inhibition of HIF-1α under hypoxic conditions in human lung adenocarcinoma A549 cells. Int. J. Mol. Sci. 2013, 14, 7273–7285. [Google Scholar] [CrossRef]
  84. Wang, Y.; Qiu, Q.; Shen, J.J.; Li, D.D.; Jiang, X.J.; Si, S.Y.; Shao, R.G.; Wang, Z. Cardiac glycosides induce autophagy in human non-small cell lung cancer cells through regulation of dual signaling pathways. Int. J. Biochem. Cell Biol. 2012, 44, 1813–1824. [Google Scholar] [CrossRef]
  85. Kherallah, R.Y.; Khawaja, M.; Olson, M.; Angiolillo, D.; Birnbaum, Y. Cilostazol: A Review of Basic Mechanisms and Clinical Uses. Cardiovasc. Drugs Ther. 2022, 36, 777–792. [Google Scholar] [CrossRef]
  86. Ikeda, Y.; Matsumata, T.; Takenaka, K.; Yamagata, M.; Sugimachi, K. Effects of doxorubicin and/or cilostazol on cancer cells during liver regeneration after two-thirds hepatectomy in rats. Oncology 1998, 55, 354–356. [Google Scholar] [CrossRef]
  87. Murata, K.; Kameyama, M.; Fukui, F.; Ohigashi, H.; Hiratsuka, M.; Sasaki, Y.; Kabuto, T.; Mukai, M.; Mammoto, T.; Akedo, H.; et al. Phosphodiesterase type III inhibitor, cilostazol, inhibits colon cancer cell motility. Clin. Exp. Metastasis 1999, 17, 525–530. [Google Scholar] [CrossRef]
  88. Sim, K.H.; Shu, M.-S.; Kim, S.; Kim, J.-Y.; Choi, B.-H.; Lee, Y.J. Cilostazol Induces Apoptosis and Inhibits Proliferation of Hepatocellular Carcinoma Cells by Activating AMPK. Biotechnol. Bioprocess. Eng. 2021, 26, 776–785. [Google Scholar] [CrossRef]
  89. Feneck, R. Phosphodiesterase inhibitors and the cardiovascular system. Contin. Educ. Anaesth. Crit. Care Pain. 2007, 7, 203–207. [Google Scholar] [CrossRef]
  90. Maurice, D.H.; Ke, H.; Ahmad, F.; Wang, Y.; Chung, J.; Manganiello, V.C. Advances in targeting cyclic nucleotide phosphodiesterases. Nat. Rev. Drug Discov. 2014, 13, 290–314. [Google Scholar] [CrossRef] [PubMed]
  91. Begum, N.; Shen, W.; Manganiello, V. Role of PDE3A in regulation of cell cycle progression in mouse vascular smooth muscle cells and oocytes: Implications in cardiovascular diseases and infertility. Curr. Opin. Pharmacol. 2011, 11, 725–729. [Google Scholar] [CrossRef] [PubMed]
  92. Francis, S.H.; Corbin, J.D. Cyclic GMP Phosphodiesterases. In Encyclopedia of Biological Chemistry, 2nd ed.; Lennarz, W.J., Lane, M.D., Eds.; Academic Press: Waltham, MA, USA, 2013; pp. 567–573. [Google Scholar]
  93. Zimmerman, N.P.; Roy, I.; Hauser, A.D.; Wilson, J.M.; Williams, C.L.; Dwinell, M.B. Cyclic AMP regulates the migration and invasion potential of human pancreatic cancer cells. Mol. Carcinog. 2015, 54, 203–215. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, H.; Kong, Q.; Wang, J.; Jiang, Y.; Hua, H. Complex roles of cAMP-PKA-CREB signaling in cancer. Exp. Hematol. Oncol. 2020, 9, 32. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, L.; Zhang, J.J.; Huang, X.Y. cAMP inhibits cell migration by interfering with Rac-induced lamellipodium formation. J. Biol. Chem. 2008, 283, 13799–13805. [Google Scholar] [CrossRef] [PubMed]
  96. Chin, K.V.; Yang, W.L.; Ravatn, R.; Kita, T.; Reitman, E.; Vettori, D.; Cvijic, M.E.; Shin, M.; Iacono, L. Reinventing the wheel of cyclic AMP: Novel mechanisms of cAMP signaling. Ann. N. Y Acad. Sci. 2002, 968, 49–64. [Google Scholar] [CrossRef] [PubMed]
  97. Chiaradonna, F.; Balestrieri, C.; Gaglio, D.; Vanoni, M. RAS and PKA pathways in cancer: New insight from transcriptional analysis. Front. Biosci. 2008, 13, 5257–5278. [Google Scholar] [CrossRef]
  98. McCubrey, J.A.; Steelman, L.S.; Chappell, W.H.; Abrams, S.L.; Wong, E.W.; Chang, F.; Lehmann, B.; Terrian, D.M.; Milella, M.; Tafuri, A.; et al. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. Biochim. Biophys. Acta 2007, 1773, 1263–1284. [Google Scholar] [CrossRef]
  99. Rangarajan, S.; Enserink, J.M.; Kuiperij, H.B.; de Rooij, J.; Price, L.S.; Schwede, F.; Bos, J.L. Cyclic AMP induces integrin-mediated cell adhesion through Epac and Rap1 upon stimulation of the beta 2-adrenergic receptor. J. Cell Biol. 2003, 160, 487–493. [Google Scholar] [CrossRef] [PubMed]
  100. Enserink, J.M.; Price, L.S.; Methi, T.; Mahic, M.; Sonnenberg, A.; Bos, J.L.; Taskén, K. The cAMP-Epac-Rap1 pathway regulates cell spreading and cell adhesion to laminin-5 through the alpha3beta1 integrin but not the alpha6beta4 integrin. J. Biol. Chem. 2004, 279, 44889–44896. [Google Scholar] [CrossRef] [PubMed]
  101. Kooistra, M.R.; Corada, M.; Dejana, E.; Bos, J.L. Epac1 regulates integrity of endothelial cell junctions through VE-cadherin. FEBS Lett. 2005, 579, 4966–4972. [Google Scholar] [CrossRef] [PubMed]
  102. Cullere, X.; Shaw, S.K.; Andersson, L.; Hirahashi, J.; Luscinskas, F.W.; Mayadas, T.N. Regulation of vascular endothelial barrier function by Epac, a cAMP-activated exchange factor for Rap GTPase. Blood 2005, 105, 1950–1955. [Google Scholar] [CrossRef] [PubMed]
  103. Kiermayer, S.; Biondi, R.M.; Imig, J.; Plotz, G.; Haupenthal, J.; Zeuzem, S.; Piiper, A. Epac activation converts cAMP from a proliferative into a differentiation signal in PC12 cells. Mol. Biol. Cell 2005, 16, 5639–5648. [Google Scholar] [CrossRef] [PubMed]
  104. Grandoch, M.; Rose, A.; ter Braak, M.; Jendrossek, V.; Rübben, H.; Fischer, J.W.; Schmidt, M.; Weber, A.A. Epac inhibits migration and proliferation of human prostate carcinoma cells. Br. J. Cancer 2009, 101, 2038–2042. [Google Scholar] [CrossRef] [PubMed]
  105. Kumar, N.; Prasad, P.; Jash, E.; Saini, M.; Husain, A.; Goldman, A.; Sehrawat, S. Insights into exchange factor directly activated by cAMP (EPAC) as potential target for cancer treatment. Mol. Cell Biochem. 2018, 447, 77–92. [Google Scholar] [CrossRef] [PubMed]
  106. Franco, R.; Bortner, C.D.; Cidlowski, J.A. Potential roles of electrogenic ion transport and plasma membrane depolarization in apoptosis. J. Membr. Biol. 2006, 209, 43–58. [Google Scholar] [CrossRef] [PubMed]
  107. Bortner, C.D.; Sifre, M.I.; Cidlowski, J.A. Cationic gradient reversal and cytoskeleton-independent volume regulatory pathways define an early stage of apoptosis. J. Biol. Chem. 2008, 283, 7219–7229. [Google Scholar] [CrossRef]
  108. Bortner, C.D.; Cidlowski, J.A. Uncoupling cell shrinkage from apoptosis reveals that Na+ influx is required for volume loss during programmed cell death. J. Biol. Chem. 2003, 278, 39176–39184. [Google Scholar] [CrossRef]
  109. Yurinskaya, V.; Goryachaya, T.; Guzhova, I.; Moshkov, A.; Rozanov, Y.; Sakuta, G.; Shirokova, A.; Shumilina, E.; Vassilieva, I.; Lang, F.; et al. Potassium and sodium balance in U937 cells during apoptosis with and without cell shrinkage. Cell Physiol. Biochem. 2005, 16, 155–162. [Google Scholar] [CrossRef] [PubMed]
  110. Hirst, D.G.; Robson, T. Nitric oxide physiology and pathology. Methods Mol. Biol. 2011, 704, 1–13. [Google Scholar] [CrossRef] [PubMed]
  111. Papapetropoulos, A.; García-Cardeña, G.; Madri, J.A.; Sessa, W.C. Nitric oxide production contributes to the angiogenic properties of vascular endothelial growth factor in human endothelial cells. J. Clin. Investig. 1997, 100, 3131–3139. [Google Scholar] [CrossRef] [PubMed]
  112. Kamm, A.; Przychodzen, P.; Kuban-Jankowska, A.; Jacewicz, D.; Dabrowska, A.M.; Nussberger, S.; Wozniak, M.; Gorska-Ponikowska, M. Nitric oxide and its derivatives in the cancer battlefield. Nitric Oxide 2019, 93, 102–114. [Google Scholar] [CrossRef] [PubMed]
  113. Choudhari, S.K.; Chaudhary, M.; Bagde, S.; Gadbail, A.R.; Joshi, V. Nitric oxide and cancer: A review. World J. Surg. Oncol. 2013, 11, 118. [Google Scholar] [CrossRef] [PubMed]
  114. Forrester, K.; Ambs, S.; Lupold, S.E.; Kapust, R.B.; Spillare, E.A.; Weinberg, W.C.; Felley-Bosco, E.; Wang, X.W.; Geller, D.A.; Tzeng, E.; et al. Nitric oxide-induced p53 accumulation and regulation of inducible nitric oxide synthase expression by wild-type p53. Proc. Natl. Acad. Sci. USA 1996, 93, 2442–2447. [Google Scholar] [CrossRef] [PubMed]
  115. Falke, D.; Fisher, M.; Ye, D.; Juliano, R.L. Design of artificial transcription factors to selectively regulate the pro-apoptotic bax gene. Nucleic Acids Res. 2003, 31, e10. [Google Scholar] [CrossRef] [PubMed]
  116. Vince, J.E.; De Nardo, D.; Gao, W.; Vince, A.J.; Hall, C.; McArthur, K.; Simpson, D.; Vijayaraj, S.; Lindqvist, L.M.; Bouillet, P.; et al. The Mitochondrial Apoptotic Effectors BAX/BAK Activate Caspase-3 and -7 to Trigger NLRP3 Inflammasome and Caspase-8 Driven IL-1β Activation. Cell Rep. 2018, 25, 2339–2353.e2334. [Google Scholar] [CrossRef] [PubMed]
  117. Chandra, D.; Choy, G.; Deng, X.; Bhatia, B.; Daniel, P.; Tang, D.G. Association of active caspase 8 with the mitochondrial membrane during apoptosis: Potential roles in cleaving BAP31 and caspase 3 and mediating mitochondrion-endoplasmic reticulum cross talk in etoposide-induced cell death. Mol. Cell Biol. 2004, 24, 6592–6607. [Google Scholar] [CrossRef]
  118. Nakamura, H.; Takada, K. Reactive oxygen species in cancer: Current findings and future directions. Cancer Sci. 2021, 112, 3945–3952. [Google Scholar] [CrossRef]
  119. Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef] [PubMed]
  120. Liou, G.Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef] [PubMed]
  121. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed]
  122. Milkovic, L.; Cipak Gasparovic, A.; Cindric, M.; Mouthuy, P.A.; Zarkovic, N. Short Overview of ROS as Cell Function Regulators and Their Implications in Therapy Concepts. Cells 2019, 8, 793. [Google Scholar] [CrossRef] [PubMed]
  123. Villalpando-Rodriguez, G.E.; Gibson, S.B. Reactive Oxygen Species (ROS) Regulates Different Types of Cell Death by Acting as a Rheostat. Oxidative Med. Cell. Longev. 2021, 2021, 9912436. [Google Scholar] [CrossRef] [PubMed]
  124. Waris, G.; Ahsan, H. Reactive oxygen species: Role in the development of cancer and various chronic conditions. J. Carcinog. 2006, 5, 14. [Google Scholar] [CrossRef]
  125. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxid. Med. Cell Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef]
  126. Lee, J.D.; Cai, Q.; Shu, X.O.; Nechuta, S.J. The Role of Biomarkers of Oxidative Stress in Breast Cancer Risk and Prognosis: A Systematic Review of the Epidemiologic Literature. J. Womens Health 2017, 26, 467–482. [Google Scholar] [CrossRef]
  127. Zhang, L.; Li, L.; Gao, G.; Wei, G.; Zheng, Y.; Wang, C.; Gao, N.; Zhao, Y.; Deng, J.; Chen, H.; et al. Elevation of GPRC5A expression in colorectal cancer promotes tumor progression through VNN-1 induced oxidative stress. Int. J. Cancer 2017, 140, 2734–2747. [Google Scholar] [CrossRef] [PubMed]
  128. Saijo, H.; Hirohashi, Y.; Torigoe, T.; Horibe, R.; Takaya, A.; Murai, A.; Kubo, T.; Kajiwara, T.; Tanaka, T.; Shionoya, Y.; et al. Plasticity of lung cancer stem-like cells is regulated by the transcription factor HOXA5 that is induced by oxidative stress. Oncotarget 2016, 7, 50043–50056. [Google Scholar] [CrossRef]
  129. Wang, Z.; Li, Z.; Ye, Y.; Xie, L.; Li, W. Oxidative Stress and Liver Cancer: Etiology and Therapeutic Targets. Oxid. Med. Cell Longev. 2016, 2016, 7891574. [Google Scholar] [CrossRef]
  130. Oh, B.; Figtree, G.; Costa, D.; Eade, T.; Hruby, G.; Lim, S.; Elfiky, A.; Martine, N.; Rosenthal, D.; Clarke, S.; et al. Oxidative stress in prostate cancer patients: A systematic review of case control studies. Prostate Int. 2016, 4, 71–87. [Google Scholar] [CrossRef]
  131. Saed, G.M.; Diamond, M.P.; Fletcher, N.M. Updates of the role of oxidative stress in the pathogenesis of ovarian cancer. Gynecol. Oncol. 2017, 145, 595–602. [Google Scholar] [CrossRef] [PubMed]
  132. Jaroonwitchawan, T.; Chaicharoenaudomrung, N.; Namkaew, J.; Noisa, P. Curcumin attenuates paraquat-induced cell death in human neuroblastoma cells through modulating oxidative stress and autophagy. Neurosci. Lett. 2017, 636, 40–47. [Google Scholar] [CrossRef]
  133. Chen, X.; Guo, C.; Kong, J. Oxidative stress in neurodegenerative diseases. Neural Regen. Res. 2012, 7, 376–385. [Google Scholar] [CrossRef]
  134. Bailo, P.S.; Martín, E.L.; Calmarza, P.; Breva, S.M.; Gómez, A.B.; Giráldez, A.P.; Callau, J.J.S.-P.; Santamaría, J.M.V.; Khialani, A.D.; Micó, C.C.; et al. The role of oxidative stress in neurodegenerative diseases and potential antioxidant therapies. Adv. Lab. Med. / Av. En Med. De Lab. 2022, 3, 342–350. [Google Scholar] [CrossRef] [PubMed]
  135. Dubois-Deruy, E.; Peugnet, V.; Turkieh, A.; Pinet, F. Oxidative Stress in Cardiovascular Diseases. Antioxidants 2020, 9, 864. [Google Scholar] [CrossRef]
  136. Senoner, T.; Dichtl, W. Oxidative Stress in Cardiovascular Diseases: Still a Therapeutic Target? Nutrients 2019, 11, 2090. [Google Scholar] [CrossRef]
  137. Khan, A.Q.; Kuttikrishnan, S.; Siveen, K.S.; Prabhu, K.S.; Shanmugakonar, M.; Al-Naemi, H.A.; Haris, M.; Dermime, S.; Uddin, S. RAS-mediated oncogenic signaling pathways in human malignancies. Semin. Cancer Biol. 2019, 54, 1–13. [Google Scholar] [CrossRef]
  138. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616. [Google Scholar] [CrossRef]
  139. Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [PubMed]
  140. Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.A.; Sethi, G. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules 2019, 9, 735. [Google Scholar] [CrossRef] [PubMed]
  141. Weinberg, F.; Ramnath, N.; Nagrath, D. Reactive Oxygen Species in the Tumor Microenvironment: An Overview. Cancers 2019, 11, 1191. [Google Scholar] [CrossRef] [PubMed]
  142. Rodic, S.; Vincent, M.D. Reactive oxygen species (ROS) are a key determinant of cancer’s metabolic phenotype. Int. J. Cancer 2018, 142, 440–448. [Google Scholar] [CrossRef] [PubMed]
  143. Kim, J.; Kim, J.; Bae, J.S. ROS homeostasis and metabolism: A critical liaison for cancer therapy. Exp. Mol. Med. 2016, 48, e269. [Google Scholar] [CrossRef] [PubMed]
  144. Muri, J.; Kopf, M. Redox regulation of immunometabolism. Nat. Rev. Immunol. 2021, 21, 363–381. [Google Scholar] [CrossRef] [PubMed]
  145. Fox, D.B.; Garcia, N.M.G.; McKinney, B.J.; Lupo, R.; Noteware, L.C.; Newcomb, R.; Liu, J.; Locasale, J.W.; Hirschey, M.D.; Alvarez, J.V. NRF2 activation promotes the recurrence of dormant tumour cells through regulation of redox and nucleotide metabolism. Nat. Metab. 2020, 2, 318–334. [Google Scholar] [CrossRef] [PubMed]
  146. Kirkpatrick, D.L.; Powis, G. Clinically Evaluated Cancer Drugs Inhibiting Redox Signaling. Antioxid. Redox Signal 2017, 26, 262–273. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, Y.; Qi, H.; Liu, Y.; Duan, C.; Liu, X.; Xia, T.; Chen, D.; Piao, H.L.; Liu, H.X. The double-edged roles of ROS in cancer prevention and therapy. Theranostics 2021, 11, 4839–4857. [Google Scholar] [CrossRef]
  148. Kirtonia, A.; Sethi, G.; Garg, M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell Mol. Life Sci. 2020, 77, 4459–4483. [Google Scholar] [CrossRef]
  149. Ralph, S.J.; Nozuhur, S.; RA, A.L.; Rodríguez-Enríquez, S.; Moreno-Sánchez, R. Repurposing drugs as pro-oxidant redox modifiers to eliminate cancer stem cells and improve the treatment of advanced stage cancers. Med. Res. Rev. 2019, 39, 2397–2426. [Google Scholar] [CrossRef] [PubMed]
  150. Mannino, F.; Urzì Brancati, V.; Lauro, R.; Pirrotta, I.; Rottura, M.; Irrera, N.; Cavallini, G.M.; Pallio, G.; Gitto, E.; Manti, S. Levosimendan and Dobutamin Attenuate LPS-Induced Inflammation in Microglia by Inhibiting the NF-κB Pathway and NLRP3 Inflammasome Activation via Nrf2/HO-1 Signalling. Biomedicines 2024, 12, 1009. [Google Scholar] [CrossRef] [PubMed]
  151. Zhao, P.; Han, S.N.; Arumugam, S.; Yousaf, M.N.; Qin, Y.; Jiang, J.X.; Torok, N.J.; Chen, Y.; Mankash, M.S.; Liu, J.; et al. Digoxin improves steatohepatitis with differential involvement of liver cell subsets in mice through inhibition of PKM2 transactivation. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G387–G397. [Google Scholar] [CrossRef] [PubMed]
  152. Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef] [PubMed]
  153. Weydert, C.J.; Cullen, J.J. Measurement of superoxide dismutase, catalase and glutathione peroxidase in cultured cells and tissue. Nat. Protoc. 2010, 5, 51–66. [Google Scholar] [CrossRef] [PubMed]
  154. Zhou, X.; An, B.; Lin, Y.; Ni, Y.; Zhao, X.; Liang, X. Molecular mechanisms of ROS-modulated cancer chemoresistance and therapeutic strategies. Biomed. Pharmacother. 2023, 165, 115036. [Google Scholar] [CrossRef]
  155. Chen, X.; Song, M.; Zhang, B.; Zhang, Y. Reactive Oxygen Species Regulate T Cell Immune Response in the Tumor Microenvironment. Oxid. Med. Cell Longev. 2016, 2016, 1580967. [Google Scholar] [CrossRef]
  156. Begley, C.G.; Ashton, M.; Baell, J.; Bettess, M.; Brown, M.P.; Carter, B.; Charman, W.N.; Davis, C.; Fisher, S.; Frazer, I.; et al. Drug repurposing: Misconceptions, challenges, and opportunities for academic researchers. Sci. Transl. Med. 2021, 13, eabd5524. [Google Scholar] [CrossRef]
Figure 1. Mechanism of action of positive inotropic agents. β-Agonist compounds bind to β receptors, triggering the production of cyclic AMP (cAMP) from AMP via adenylate cyclase. This cAMP then activates protein kinase A (PKa), which in turn stimulates the release of calcium ions (Ca2+) from the sarcoplasmic reticulum into the cell’s cytosol through phosphorylated ryanodine receptors. The presence of Ca2+ enables troponin C (Tnc) to bind effectively, leading to the activation of actin-myosin filament interactions and ultimately muscle contraction. Phosphodiesterase (PDE) inhibitors also play a role in increasing cAMP levels by preventing its breakdown. Ca2+ sensitizers enhance the responsiveness of Tnc to Ca2+, thus augmenting the strength of muscle contraction. Levosimendan binds to Tnc during systole, increasing the sensitivity of myofilaments to Ca2+ levels. Cardiac myosin activators boost the activity of myofibril ATPase, intensifying the contractile force of cardiac cells without necessitating more ATP molecules for contraction. Cardiac glycosides function by inhibiting Na+/K+-ATPase. This inhibition elevates the activity of the sarcoendoplasmic reticulum calcium ATPase pump, facilitating the reuptake of Ca2+ by the sarcoplasmic reticulum and enhancing cardiac muscle contraction.
Figure 1. Mechanism of action of positive inotropic agents. β-Agonist compounds bind to β receptors, triggering the production of cyclic AMP (cAMP) from AMP via adenylate cyclase. This cAMP then activates protein kinase A (PKa), which in turn stimulates the release of calcium ions (Ca2+) from the sarcoplasmic reticulum into the cell’s cytosol through phosphorylated ryanodine receptors. The presence of Ca2+ enables troponin C (Tnc) to bind effectively, leading to the activation of actin-myosin filament interactions and ultimately muscle contraction. Phosphodiesterase (PDE) inhibitors also play a role in increasing cAMP levels by preventing its breakdown. Ca2+ sensitizers enhance the responsiveness of Tnc to Ca2+, thus augmenting the strength of muscle contraction. Levosimendan binds to Tnc during systole, increasing the sensitivity of myofilaments to Ca2+ levels. Cardiac myosin activators boost the activity of myofibril ATPase, intensifying the contractile force of cardiac cells without necessitating more ATP molecules for contraction. Cardiac glycosides function by inhibiting Na+/K+-ATPase. This inhibition elevates the activity of the sarcoendoplasmic reticulum calcium ATPase pump, facilitating the reuptake of Ca2+ by the sarcoplasmic reticulum and enhancing cardiac muscle contraction.
Targets 02 00009 g001
Figure 2. Schematic representation of the proposed mechanisms by which cilostazol exerts anticancer effects in HCC cells. Cilostazol triggers cell cycle arrest and apoptosis in HCC cells, leading to reduced cell viability and proliferation through the activation of AMPK signaling. Additionally, AMPK activation suppresses the pro-proliferative AKT and ERK signaling pathways in HCC cells.
Figure 2. Schematic representation of the proposed mechanisms by which cilostazol exerts anticancer effects in HCC cells. Cilostazol triggers cell cycle arrest and apoptosis in HCC cells, leading to reduced cell viability and proliferation through the activation of AMPK signaling. Additionally, AMPK activation suppresses the pro-proliferative AKT and ERK signaling pathways in HCC cells.
Targets 02 00009 g002
Figure 3. Dual effect of nitric oxide (NO) on tumor cells. High levels induce cell death, while low levels promote survival.
Figure 3. Dual effect of nitric oxide (NO) on tumor cells. High levels induce cell death, while low levels promote survival.
Targets 02 00009 g003
Table 1. Examples of successfully repurposed drugs.
Table 1. Examples of successfully repurposed drugs.
Drug NameOriginal IndicationNew IndicationReferences
VerapamilHeart rhythm problemsFungal biofilms[15]
SildenafilAnginaErectile dysfunction[16]
CarboplatinAntitumorFungal biofilms[15]
AmilorideHypertensionSecondary progressive multiple sclerosis (SPMS)[17]
MinoxidilHypertensionHair loss[18]
AspirinAnalgesiaColorectal cancer[19]
ThalidomideMorning sicknessMultiple myeloma[20]
ZidovudineCancerHIV[21]
RituximabCancerRheumatoid arthritis[22]
RaloxifeneOsteoporosisBreast cancer[23]
Table 2. Common mechanisms of action of positive inotropic agents in cancer.
Table 2. Common mechanisms of action of positive inotropic agents in cancer.
CategoryDrugIndicationMechanism of Action
Cardiac glycosidesDigoxinMild-to-moderate heart failure, increased myocardial contraction, and maintained control ventricular rateNa-K ATPase enzyme inhibition
Phosphodiesterase
III (PDE3) inhibitor
EnoximoneCongestive heart failurePDE3 inhibition
Stimulates NO production
Increases cAMP levels
MilrinoneAcute decompensated heart failurePDE3 inhibition
Increases cAMP levels
AmrinoneCongestive heart failurePDE3 inhibition
Increases cAMP levels
β-agonistsDobutamineCardiac decompensationIncreases cAMP levels
DopamineHypotensionIncreases cAMP levels
IsoetharineAsthmaIncreases cAMP levels
RitodrinePremature laborIncreases cAMP levels
TerbutalineAsthma and premature laborIncreases cAMP levels
Calcium sensitizerLevosimendanChronic heart failurePDE3 inhibition
Stimulates NO production
Decreases ROS levels
Increases cAMP levels
cAMP = cyclic adenosine monophosphate; NO = nitric oxide; ROS = reactive oxygen species.
Table 3. Positive inotropic agents used in the manuscript.
Table 3. Positive inotropic agents used in the manuscript.
DrugType of CancerAnticancer ActivityDosesMaximum Dose Used (Cardiovascular Diseases)References
DopamineBreast and colonInhibits angiogenesis and tumor growth50 mg/kg/day50 µg/kg/min[62]
DobutamineGastricInhibits cell growth, migration, cell colony formation, and cell invasion, arrests the cell cycle at the G1 or S phase, and increases the rate of apoptosis30 μmol/L40 µg/kg/min[71]
BoneInhibits cell growth, migration, and cell invasion, augments cell apoptosis, and arrests the cell cycle in the G2 or M phase10 µM[38]
DigoxinNSCLCReduces the cell viability, increases DNA damage by promoting ROS generation, and inhibitis both DNA double-strand break (DSB) and single-strand break (SSB) repair0.2 µM–
1.0 mg/kg/day
0.75–1.5 mg/day[31]
CilostazolColonSuppress migration50 µM200 mg/day[87]
LiverInhibits proliferation, induces apoptosis, induces G0 and G1 cell cycle arrest, and decreases the expression of cyclin D1 and nuclear antigen in proliferating cells100 µM[88]
LevosimendanBladder and prostateInhibits cell migration, cell colony formation, and proliferation100 µM6–12 µg/kg/10 minOur Lab
Table 4. Standard chemotherapy drugs for cancer treatment. Adapted from emedicine.medscape.com.
Table 4. Standard chemotherapy drugs for cancer treatment. Adapted from emedicine.medscape.com.
DrugType of CancerDoses
DoxorubicinBreast, bone, and liver60–75 mg/m2/d
5-FUColon and gastric500–2600 mg/m2/d
Cisplatin and pemetrexedNSCLC75 mg/m2/d + 500 mg/m2/d
Gemcitabine and cisplatinBladder1000 mg/m2/d + 70 mg/m2/d
Docetaxel and prednisoneProstate75 mg/m2/d + 10 mg/d
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

Ribeiro, E.; Vale, N. Positive Inotropic Agents in Cancer Therapy: Exploring Potential Anti-Tumor Effects. Targets 2024, 2, 137-156. https://doi.org/10.3390/targets2020009

AMA Style

Ribeiro E, Vale N. Positive Inotropic Agents in Cancer Therapy: Exploring Potential Anti-Tumor Effects. Targets. 2024; 2(2):137-156. https://doi.org/10.3390/targets2020009

Chicago/Turabian Style

Ribeiro, Eduarda, and Nuno Vale. 2024. "Positive Inotropic Agents in Cancer Therapy: Exploring Potential Anti-Tumor Effects" Targets 2, no. 2: 137-156. https://doi.org/10.3390/targets2020009

APA Style

Ribeiro, E., & Vale, N. (2024). Positive Inotropic Agents in Cancer Therapy: Exploring Potential Anti-Tumor Effects. Targets, 2(2), 137-156. https://doi.org/10.3390/targets2020009

Article Metrics

Back to TopTop