Next Article in Journal
Role of Trientine in Hypertrophic Cardiomyopathy: A Review of Mechanistic Aspects
Previous Article in Journal
Dilemmas in the Choice of Adequate Therapeutic Treatment in Patients with Acute Pulmonary Embolism—From Modern Recommendations to Clinical Application
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Montelukast and Acute Coronary Syndrome: The Endowed Drug

1
Internal Medicine, Endocrinology and Diabetes, Department of Medicine, College of Medicine, Aljouf University, Sakaka 72388, Saudi Arabia
2
Department of Pharmacology and Toxicology, College of Medicine, Al-Mustansiriyah University, Baghdad P.O. Box 14132, Iraq
3
Smartox Biotechnology, 6 Rue des Platanes, 38120 Saint-Egrève, France
4
L’institut du Thorax, INSERM, CNRS, Université de Nantes, 44007 Nantes, France
5
LabEx «Ion Channels, Science & Therapeutics», Université de Nice Sophia-Antipolis, 06560 Valbonne, France
6
Institut de Neurophysiopathologie (INP), Faculté des Sciences Médicales et Paramédicales, Aix-Marseille Université, CNRS UMR 7051, 27 Bd Jean Moulin, 13005 Marseille, France
7
Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh 51744, Egypt
8
Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2022, 15(9), 1147; https://doi.org/10.3390/ph15091147
Submission received: 18 August 2022 / Revised: 6 September 2022 / Accepted: 9 September 2022 / Published: 14 September 2022
(This article belongs to the Section Pharmacology)

Abstract

:
Acute coronary syndrome (ACS) is a set of signs and symptoms caused by a reduction of coronary blood flow with subsequent myocardial ischemia. ACS is associated with activation of the leukotriene (LT) pathway with subsequent releases of various LTs, including LTB4, LTC4, and LTD4, which cause inflammatory changes and induction of immunothrombosis. LTs through cysteine leukotriene (CysLT) induce activation of platelets and clotting factors with succeeding coronary thrombosis. CysLT receptor (CysLTR) antagonists such as montelukast (MK) may reduce the risk of the development of ACS and associated complications through suppression of the activation of platelet and clotting factors. Thus, this critical review aimed to elucidate the possible protective role of MK in the management of ACS. The LT pathway is implicated in the pathogenesis of atherosclerosis, cardiac hypertrophy, and heart failure. Inhibition of the LT pathway and CysL1TR by MK might be effective in preventing cardiovascular complications. MK could be an effective novel therapy in the management of ACS through inhibition of pro-inflammatory CysLT1R and modulation of inflammatory signaling pathways. MK can attenuate thrombotic events by inhibiting platelet activation and clotting factors that are activated during the development of ACS. In conclusion, MK could be an effective agent in reducing the severity of ACS and associated complications. Experimental, preclinical, and clinical studies are recommended to confirm the potential therapeutic of MK in the management of ACS.

Graphical Abstract

1. Introduction

Acute coronary syndrome (ACS) is a set of signs and symptoms caused by a reduction of coronary blood flow with subsequent myocardial ischemia [1]. According to the electrocardiographic (ECG) changes and duration of symptoms, ACS is divided into unstable angina, which represents 38%; ST-elevation myocardial infarction (STEMI) representing 30%, and non-ST-elevation myocardial infarction (NSTEMI) representing 25%. Unstable angina of ACS is differentiated from stable angina, as it occurs suddenly at rest with little response to treatment [2].
The cardinal clinical features of ACS are chest pain, dyspnea, nausea, vomiting, sweating, and tachycardia [3]. ACS is mainly caused by coronary thrombosis, coronary vasospasm, aortic valve stenosis, severe anemia, and pulmonary hypertension [4]. The underlying pathogenic mechanisms of ACS are related to rupture of atheroma in 60% and atheroma erosion in 30%. Most of the atheroma rupture induces the development of STEMI, while the development of atheroma erosion induces propagation of NSTEMI [5].
Plague rupture provokes in situ inflammatory reactions and thrombosis with subsequent ischemic-reperfusion injury (IRI). Thrombolytic therapy is indicated for STEMI, while nitroglycerin, antiplatelet, low-molecular-weight heparin (LMWH) such as enoxaparin, and heparin-like drugs such as fondaparinux are effective in this state [6,7].
It has been reported that leukotriene receptor (LT) receptor antagonists such as montelukast (MK) may have potential beneficial effects in the cardiovascular field [8]. Thus, this critical review aimed to elucidate the possible protective role of MK in the management of ACS.

2. Leukotriene Pathway

The LT name was initially described in 1979 by Swedish Bengt Samuelsson as a slow-reaction, smooth-muscle-stimulating substance that was introduced by Feldberg and Kellaway between 1938 and 1940 [9]. LTs are inflammatory eicosanoid mediators generated by leukocytes through oxidation of arachidonic acid (AA) and eicosapentaenoic acid (EPA) by the action of 5-lipoxygenase (5-LO) [9]. LTs use lipid signaling to express and convey information in autocrine and paracrine patterns to control immune functions. Usually, the production of LTs is correlated with the release of prostaglandins and histamines [10]. By the action of 5LO or 5LO-activating protein (FLAP), the AA is converted to LTA4, which is converted through LTA4 hydrolase and LTC4 synthase to LTB4 and cysteinleukotriene (CysLT), respectively. LTB4 is inhibited by LY-293111 a specific inhibitor of the LTB4 receptor, while MK, pranlukast, and zafirlukast block CysLT receptor (CysLTR) type 1 [11] (Figure 1).
Thus, there are two types of LTs, namely LTB4 and CysLT, which include LTC4, LTE4, and LTD4. CysLTs are involved in the development of allergy and anaphylaxis. Specifically, LTB4 induces recruitment of neutrophils with induction of tissue injury and release of pro-inflammatory cytokines. Therefore, LTB4 antagonists inhibit the development and propagation of neutrophil-mediated disorders [12,13]. LTA4 is mainly converted to LTB4 and has few inflammatory effects. LTC4 is mainly expressed by eosinophil and mast cells, which are converted outside the cells to form LTD4 and LTE4, which have inflammatory effects. Moreover, LTG4 is regarded as an active metabolite of LTE4 that has unknown biological effects [14,15].
There are two main types of CysLT receptors, which are CysLTR1 and CysLTR2. CysLTR1 is a G-protein coupled receptor that is mainly activated by LTD4 and LTC4, causing airway bronchoconstriction and increasing vascular permeability, the influx of neutrophils and eosinophils, fibrosis and collagen deposition, and membrane hypertrophy of respiratory epithelium [16]. Additionally, activation of CysLTR1 is associated with disruption of the blood–brain barrier (BBB) with induction of neuroinflammation and convulsion. Therefore, CysLTR1 antagonists could be effective against the development of Parkinson’s disease, Alzheimer’s disease, encephalomyelitis, and multiple sclerosis [17]. CysLTR1 is inhibited by MK, pranlukast, and zafirlukast [17]. CysLTR2 is activated by LTD4 and LTC4, and it is involved in the development of hypersensitivity and allergic reactions [18,19]. CysLTR2 is chiefly expressed in the platelets, eosinophils, macrophages, and mast cells. Furthermore, CysLTR2 is highly expressed in the heart, adrenal gland, endothelial cells, smooth muscle cells, and airway epithelial cells [18]. MK and other CysLTs only block CysLTR1; however, gemilukast inhibits both CysLTR1 and CysLTR2, which can be effective in the management of asthma [20]. Other types of LT receptors such as GPR99, which is called oxoglutarate or CysLTR3, are stimulated by LTE4 and α-ketoglutarate, while GPR99 is inhibited by MK [21]. Overexpression of GPR99 is linked with the development of hypertension [21].
GPR17 is a G-protein coupled receptor stimulated by LTC4, LTD4, and uracil nucleotides. GPR17 chemical structure is intermediate between CysLTR1 and CysLTR2. GPR17 is highly distributed in the central nervous system (CNS) mainly in the microglia cells [22]. Cangrelor, which is a purinergic receptor (P2Y) antagonist, blocks GPR17 [23].
Furthermore, P2Y12 is activated following activation of CysLTR1 by LTs; therefore, inhibition of P2Y12 by a specific antagonist attenuates the CysLTR1-dependent effect of LTs. P2Y12 is a chemoreceptor stimulated by adenosine diphosphate (ADP) and belongs to G-protein coupled purinergic receptor group [24]. P2Y12 is involved in platelet aggregation and thrombosis; it is inhibited by antiplatelet drugs such as clopidogrel, prasugrel, ticagrelor, and cangrelor [24]. Notably, P2Y12 is also expressed in the vascular smooth muscle and immune cells and involved in the pathogenesis of inflammation through the promotion of the interaction between platelets and leukocytes with the release of inflammatory cytokines [24]. Kang et al. illustrated that MK attenuates bone loss through inhibition expression of P2Y12 [25]. Moreover, LTs can activate peroxisome proliferator-activated receptors (PPAR). Interestingly, zafirlukast and MK are regarded as a potent modulators of PPAR-γ [26]. LTs are intricate in the pathogenesis of asthma, dementia, and cognitive dysfunction [26].
These findings suggest that LTs through activation of LTRs and other related receptors may be involved in the pathogenesis of respiratory and systemic disorders.

3. Pharmacology of Montelukast

MK is a monocarboxylic acid with aliphatic sulfide from the quinolone family (Figure 2).
MK is an orally active CysLTR1 antagonist that blocks the action of LTD4 used in the prophylaxis of asthma and management of various inflammatory disorders [28]. Similarly, MK is indicated in the management of urticarial and allergic rhinitis [28]. MK was approved by the Food and Drug Administration (FDA) in 1998 in the USA. The use of MK is associated with the development of many adverse effects, including vomiting, nausea, skin rash, angioedema, mild fever, malaise, paresthesia, muscle cramps, and seizure [28,29]. Prolonged use of MK is linked with the development of neuropsychiatric disorders, including nightmares, insomnia, depression, anxiety, and aggression [29]. In 2009, the FDA declared that long-term use of MK was associated with increased risk of suicidal behavior [29]. In 2020, the FDA put MK in a boxed warning about the risk of neuropsychiatric disorders and advises to limit the use of MK for simple allergic reactions when safe alternatives are available [30].
MK is rapidly absorbed from the intestine with 64% bioavailability, and its absorption is not affected by food. MK plasma concentration is attained within 2–4 h with high plasma protein binding. MK has few drug–drug interactions though it inhibits liver metabolizing enzyme CYP2C8. MK and its metabolites are highly distributed and excreted by bile [31]. The lethal dose in 50% (LD50) of MK is more than 5000 mg/kg [31].

4. Montelukast and Cardiovascular Complications

The general expression of CysLTR is associated with the propagation of different cardiovascular complications [32]. CysLT2R is highly expressed in the human atrium, ventricles, apex, septum, and Purkinje cells. In addition, CysLT2R is expressed on the endothelial cells, monocytes, and smooth muscle cells, while CysLT1R is chiefly present in the macrophages and monocytes [32]. Surprisingly, only CysLT2R is expressed in coronary smooth muscles, whereas aortic smooth muscles highly expressed CysLT1R [32]. Of note, CysLTR exerts negative inotropic effects and reduces coronary blood flow without chronotropic effects [33].
MK and other CysLTR antagonists were originally developed for the management of asthma. In virtue of its anti-inflammatory property, MK could be effective in treating atherosclerosis and metabolic syndrome [34]. It has been shown that asthmatic patients on MK treatments showed a low cardiovascular risk index since MK decreases the risk of stroke, myocardial infarction, and other cardiovascular complications [8]. Of interest, MK inhibits epoxide hydrolase, which hydrolyzes beneficial epoxyeicosatrienoic acid to inactive dihydroxyeicosatrienoic acid [35]. Thus, MK and other CysLTR antagonists through activation of PPAR-γ and inhibition of epoxide hydrolase may reduce cardiovascular complications through the improvement of endothelial function and insulin sensitivity [36]. Animal model studies revealed that MK reduced the risk for the development of myocardial infarction, ischemic reperfusion injury, and atherosclerosis [37,38]. Ge et al. observed that MK had an anti-atherogenic effect by inhibiting the expression of monocyte chemoattractant protein 1 (MCP-1) in rabbits with induced carotid injury [38]. Likewise, Liu’s experimental study confirmed that MK reduced the propagation of atherosclerosis by inhibiting the expression of matrix metalloproteinases (MMPs) (Figure 3) [39]. Activation of PPAR-γ and inhibition of soluble epoxide hydrolase by MK attenuates propagation of oxidative stress-induced endothelial injury through suppression expression of AA metabolites [38]. Wang and colleagues observed that over-expression of soluble epoxide hydrolase was engaged with the development of atherosclerosis by reducing the anti-inflammatory anti-thrombotic effects of epoxyeicosatrienoic acid [40].
In the atherosclerotic lesions, the expression of 5-LO and FLAP and their metabolites, including LTB4, LTA4, and LTC4 and LTD4 and LTC4 are increased with the augmentation of the expression of CysLTR [41]. These findings suggest close relationships between the development of atherosclerosis and the activation of LT pathway, so targeting this pathway could limit the progression of the atherosclerotic process. In atherosclerotic lesions, the activated macrophages are augmented, which are the major source of 5-LO [41]. A clinical study observed that long-term treatment with MK reduced serum level of C-reactive protein (CRP) in the asthmatic patients [42]. Long-term follow-up studies proposed that MK by its anti-inflammatory effect may inhibit the advancement of atherosclerosis [43]. It has been shown that LT signaling pathway is implicated in the pathogenesis of atherosclerosis through suppression of lipid retention, accumulation of foam cells, and intimal hyperplasia with subsequent propagation of atherosclerosis [43].
Furthermore, LT increases the risk for rupture of atherosclerotic plaque through degradation of extracellular matrix [43]. For these reasons, augmentation of LTs may be correlated with the development of ischemic changes. In addition, over-expression of CysLTR in the atherosclerotic lesions increases the risk of atherosclerotic complications such as myocardial infarction, ischemic stroke, and aortic aneurysm [44]. Deletion of 5-LO in experimental mice reduced the development of atherosclerotic lesions by about 25-fold [45]. Likewise, an experimental study demonstrated that atorvastatin inhibits the advancement of atherosclerotic lesions by inhibiting the expression of CysLT1R and FLAP as well as regulating the expression of 5-LO [46].
Regarding clinical studies, Revan et al. found that genetic variation in the LT pathway was linked with the development of early atherosclerotic lesions [47]. Genetic variations in the expression of 5-LO are associated with increasing intima-media thickness and progression of atherosclerosis in a cohort study [48]. These verdicts implicated the LT pathway in the pathogenesis of atherosclerosis; hence, LT pathway inhibitors or CysLTR antagonists could be effective in preventing the development of atherosclerosis. Different studies revealed that CysLT1R antagonists such as MK and FLAP inhibitors decrease the development of atherosclerosis in mice [49,50].
In the atherosclerotic lesions, LTC4 expression and its mediator, known as multi-drug resistance protein 1 (MRP-1), were increased, causing oxidative stress injury in humans [51]. Of interest, LTC4 and LTD4 increase the expression of adhesion molecules such as P-selectin in the CysLT2R-dependent pathway, and thus, MK may be ineffective in the suppression of this pathway, as it merely blocks CysLT1R [52]. Furthermore, the induction release of pro-inflammatory cytokines and chemokines by LTD4 was not suppressed by the action of MK and other CysL1TR antagonists, as the release of inflammatory cytokines are mainly mediated by the CysL2TR [53]. Remarkably, LTD4 can induce the release of anti-inflammatory cytokine IL-10, which has a protective immunomodulatory effect against the development of atherosclerosis [53].
Moreover, overexpression of GPR99 is associated with the development of cardiac hypertrophy [54]. Nguyen et al. illustrated that MK can inhibit the pharmacological activity of GPR99, thereby limiting inflammatory progression and development of cardiac hypertrophy [55]. Further, changes in the expression of GPR107 and GPR99 could be the possible cause for the propagation and pathophysiology of hypertension in rats [21]. Moreover, the development and progression of chronic heart failure are associated with the escalation of the LT pathway [56]. Inhibition of LTB4 may reduce the inflammatory changes and progression of heart failure [56]. Suppression of LTB4 and AA metabolites decreases the risk for the development of heart failure [56].
These observations suggest that the LT pathway is implicated in the pathogenesis of atherosclerosis, cardiac hypertrophy, and heart failure. Inhibition of the LT pathway and CysL1TR by MK might be effective in preventing cardiovascular complications.

5. Montelukast and Acute Coronary Syndrome

Notably, soluble epoxide hydrolase increases the expression of fatty acid synthase in the mononuclear cells in patients with ACS [57]. A prospective study involving 65 patients with ACS compared to 65 healthy controls illustrated that serum levels of soluble epoxide hydrolase and fatty acid synthase were increased in ACS patients [57]. A study including 667 ACS patients revealed that high-soluble epoxide hydrolase serum level is correlated with the severity of ACS [58]. Suppression of soluble epoxide hydrolase attenuates cardiac ischemic-reperfusion injury in vivo [59]. In addition, inhibition of soluble epoxide hydrolase attenuates the progression of hypertension, which is the major risk factor for the development of ACS [59]. These findings suggest that inhibition of soluble epoxide hydrolase by MK could be a therapeutic strategy for preventing the development of ACS.
On the other hand, PPAR-γ agonists were reported to be effective in the reduction of ACS [60]. PPAR-γ agonists reduce the expression of pro-inflammatory cytokines and macrophage activation in the coronary bed [60]. In a case-control study, PPAR-γ serum level was reduced in ACS patients and inversely correlated with cardiac troponin compared with controls [61]. Therefore, PPAR-γ serum level could be a predictive biomarker for the development of ACS mainly myocardial infarction [61]. Single-gene polymorphism in PPAR-γ predisposes the development of ACS in the Turkish population [62]. Remarkably, expression of LTC4 and CysLT1R are increased during adipogenesis through upregulation of PPAR-γ [63]. Therefore, inhibition of CysLT1R by MK might be effective in the management of obesity-induced inflammatory disorders.
Of note, LTC4 increases intima-media thickness and calcium contents in the coronary atherosclerotic plaques [64]. In addition, hypoxic stress in ACS increases the expression of CysLT1R and LTC4 in mice [49]. Within coronary atherosclerotic lesions, CysLTs are generated under hypoxic and ischemic conditions causing augmentation of vascular reactivity and propagation of coronary vasoconstriction [49,65]. Thus, CysLTs lead to noteworthy coronary hemodynamic changes. For example, administration of LTD4 in patients with significant coronary stenosis leads to an augmentation of coronary resistance measured by coronary angiogram [66]. A previous in vitro study demonstrated that LTD4- and LTC4-induced coronary vasospasm during angiography was inhibited by MK [66]. It has been shown that the sensitivity of CysLTs to induce coronary vasospasm was increased due to the release of binding molecules, which increase the binding of CysLTs to the coronary atherosclerotic lesions [67]. Moreover, CysLT1Rs expression is increased in response to the released pro-inflammatory cytokines from coronary atherosclerotic lesions [67]. Interestingly, CysLT1R expression is higher than CysLT2R in the carotid and coronary intima [68]. In an experimental study, CysLTs promote the release of pro-inflammatory cytokines with induction expression of adhesion molecules [68].
These findings proposed that CysLTs play a critical role in the pathogenesis of ACS, and the use of CysLT1R antagonists such as MK might be of value in attenuating the severity of ACS (Figure 3).

6. Montelukast and Myocardial Infarction

It has been shown that LTs are involved in the pathogenesis of myocardial infarction. Additionally, genetic variation in 5-LO increases the risk of myocardial infarction in both animals and humans [32]. Production of CysLTs is augmented in ischemic-reperfusion injury following the development of myocardial infarction [32]. Different experimental studies illustrated that levels of CysLTs are increased after acute myocardial infarction. CysLTs induce coronary vascular resistance and increase infarct size and coronary endothelial dysfunction [69,70].
During the development of myocardial infarction, several inflammatory cells invade myocardial tissues that release LTs, which also recruit inflammatory cells and induce vascular smooth contractility, the proliferation of smooth muscle cells, and the increase of vascular permeability [71]. Gross et al.’s experimental study illustrated that production of LTs were augmented due to over-expression of 5-LO, which increases the release of pro-inflammatory cytokines [72]. A cohort study illustrated that overexpression of 5-LO increases the risk for the development of myocardial infarction [73]. Therefore, MK via inhibition of CysLT1R can reduce the severity of myocardial ischemia by inhibiting apoptosis and the production of reactive oxygen species (ROS) (Figure 3) [74].
Moreover, LTD4 plays an integral role in the development of myocardial ischemia through the induction of coronary constriction. LTD4 has been reported to be a potent coronary vasoconstrictor in the isolated heart [75]. Surprisingly, zafirlukast was shown to be ineffective against experimental myocardial ischemia since myocardial ischemia is mainly mediated by CysLT2R [76]. Notably, activation of CysLT2R results in progressive myocardial ischemia with more inflammatory changes [76]. In addition, upregulation of endothelial CysLT2R increases the risk of myocardial ischemia and apoptosis [77].
Moreover, GPR17, which is a receptor for LTD4 and LTC4, increases the risk of cardiac ischemia through interaction with cardiac CysLT1R [78]. Inhibition of GPR17 by MK reduces myocardial ischemia and fibrosis [79]. Furthermore, urinary levels of CysLTs were increased following myocardial infarction and coronary bypass surgery [79]. Another study found that rupture of coronary plaque was associated with increased urinary LTs in patients with ACS [80]. However, urinary LTs as a potential biomarker of plaque rupture or inflammatory changes during myocardial infarction need to be elucidated through further study. Many studies observed that inhibition of the LT pathway increases recovery following myocardial infarction [81,82].
Despite these findings, there are controversial studies concerning the protective effects of CysLT1R antagonists in myocardial ischemia. For example, Shekher et al. found that CysLT1R antagonists did not alter infarct size and reperfusion injury following myocardial infarction [83]. Further, coronary ligation with the development of myocardial infarction and ischemic reperfusion injury in experimental mice was not ameliorated by 5-LO inhibitors or CysLT1R antagonists [77]. However, CysLT1R antagonists may produce neutral effects [84]. Genetic variation in LTA4 leads to ethnic risk factors for the development of myocardial infarction [85]. Recently, Hori et al.’s upregulation of LTB4 receptors exacerbated inflammatory changes following myocardial infarction [86].
A systematic review involving 28 studies illustrated that CysLT1R antagonists reduce the risk of myocardial infarction, ischemic stroke, and other cardiovascular complications [87]. In addition, CysLT1R antagonists could be effective for the secondary prevention of cardiovascular diseases. The reduction of recurrent myocardial infarction in male subjects on MK therapy was significant as compared to male subjects not on MK treatment (HR = 0.65, 95% CI = 0.43–0.99) in a three-year follow-up study [87]. These findings suggest that MK may affect the expression and activity of CysLT2R, which is chiefly engaged with myocardial ischemia.
Moreover, MK attenuates lipopolysaccharide (LPS)-induced myocardial injury in a dose-dependent manner [88]. Herein, MK could have cardioprotective effects against endotoxemia-induced inflammatory changes during acute myocardial injury. This effect of MK can be attributed to its anti-inflammatory and antioxidant effects [88]. A single-center, prospective, observational study showed that patients with previous myocardial infarction had higher urinary LTs as compared with unstable angina [89].

7. Effects of Montelukast on the Inflammatory Signaling Pathway in Acute Coronary Syndrome

Different inflammatory signaling pathways are activated during the development of ACS that correlated with the severity of ACS. Mitogen-activated protein kinase (MAPK) is activated during ACS [90]. MAPK family includes extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38. MAPK family and toll-like receptor 4 (TLR4) are increased in patients with ACS compared to the controls [91]. Indolfi et al. confirmed that p38MAPK was increased in the T lymphocytes in patients with ACS [92]. In a comparative study, p38MAPK activities in T lymphocytes were not significant in patients with unstable angina compared to the healthy controls. However, it was higher in patients with ST-elevation myocardial infarction [92]. These findings confirmed that p38MAPK could be a potential biomarker to differentiate subtypes of ACS.
Likewise, nod-like receptor pyrin 3 (NLRP3) inflammasome is an inflammatory signaling pathway involved in the release of pro-inflammatory cytokines with the induction of pyroptosis [19]. It has been shown that NLRP3 inflammasome is activated in ACS mainly in acute myocardial infarction, causing hyperinflammation and cardiac fibrosis [93]. Mauro et al. revealed that the NLRP3 inflammasome is highly activated in acute myocardial infarction [94]. NLRP3 inflammasome senses intracellular signaling during tissue ischemia and provokes immune response during acute myocardial infarction [94]. In addition, coronary ischemia induces activation of NLRP3 inflammasome through ROS [95]. Activated NLRP3 inflammasome in ACS triggers inflammatory changes and cardiac dysfunction. Moreover, the mechanistic target of the rapamycin (mTOR) signaling pathway is activated during heart failure following acute myocardial infarction through the autophagic process [96]. Likewise, advanced glycation end-products (AGEPs) are triggered during ACS and contribute to the development of post-myocardial remodeling and dysfunction [97].
Therefore, targeting p38MAPK, NLRP3 inflammasome, mTOR, and AGEPs may reduce the risk of complications in patients with ACS (Figure 3). It has been illustrated that MK can inhibit both p38MAPK and NLRP3 inflammasome in different studies. It has been reported that MK inhibits microglial activation through suppression of p38MAPK signaling [98]. Further, Zhou et al. demonstrated that MK attenuated neuropathic pain by inhibiting p38MAPK and NF-κB in a rat model [99]. Updated studies showed that MK alleviates hepatotoxicity by inhibiting the NLRP3 inflammasome signaling pathway in an animal model [100,101]. Furthermore, MK and other CysLT1R antagonists have abilities to block the activation of mTOR and AGEPs in various experimental studies [102,103].
These verdicts suggest that MK may reduce the severity of ACS by reducing the activity of p38MAPK, NLRP3 inflammasome, mTOR, and AGEPs. This pleiotropic effect of MK may not be mediated by blocking CysLT1R; thus, extensive researches are warranted in this regard.
Taken together, MK could be an effective novel therapy in the management of ACS through inhibition of pro-inflammatory CysLT1R and modulation of inflammasome signaling pathways. Experimental, preclinical, and clinical studies are recommended to confirm the potential therapeutic of MK in the management of ACS.

8. Montelukast and Thrombosis in Acute Coronary Syndrome

Thrombosis plays an integral role in the pathogenesis of ACS since disruption and rupture of coronary atherosclerotic plaque expose subendothelial collagen with the release of tissue factors and pro-coagulant molecules, which activate platelet activations and fibrin formation [104]. These changes induce endothelial dysfunction and immunothrombosis with the development of coronary obstruction and subsequent ischemic-reperfusion injury in ACS [104,105]. Platelet activations trigger atheromatous complications after rupture of the coronary atheromatous plaque [104,105].
Moreover, inflammation through activation of monocytes, macrophages, and mast and T cells contributes to the pathogenesis of ACS via rupture of atherosclerotic plaque and release of pro-inflammatory cytokines [106]. Pro-inflammatory cytokines induce the expression of endothelial adhesion molecules with succeeding thrombus formation [106,107,108].
It has been shown through in silico and vitro experiments that MK had an antithrombotic effect by inhibiting factor XI (FXIa) [109]. In vivo study has also confirmed the inhibitory effect of MK on the activation of FXIa [110]. Houard and colleagues illustrated that expression of 5-LO and LTB4 are increased in the intramural thrombus [110]. Thus, LTs are engaged with the propagation of coronary thrombosis. A previous study revealed that peptido-LTs are potent agonists of clotting factors including the von Willebrand factor with induction expression of P-selectin from endothelial cells [111]. Notably, LTC4 induces the release of thromboxane A2, IL-33, and high-mobility group box 1 (HMGB1) from activated platelets, causing immunothrombosis [112,113,114]. Tang et al. observed that stimulation of GRP91 increases platelet aggregation and release of LT4 [115]. Therefore, CysLT1R inhibitors such as MK may reduce platelet activation and aggregation during the propagation of ACS and thrombotic events.
Taken together, MK has the ability to attenuate thrombotic events by inhibiting platelet activation and clotting factors, which are activated during the development of ACS (Figure 3).
The present review had many limitations, including a paucity of clinical studies that evaluate the clinical effectiveness of MK in patients with ACS. Moreover, sequential levels of LTs were not assessed in most of the clinical studies to find the association with severity and recovery.

9. Conclusions

ACS is associated with activation of LT pathway with subsequent releases of various LTs, including LTB4, LTC4, and LTD4, which cause inflammatory changes and the induction of immunothrombosis. LTs through CysLTRs induce activation of platelets and clotting factors with succeeding coronary thrombosis. CysLTR antagonists such as MK may reduce risk development of ACS and associated complications through suppression activation of platelet and clotting factors. Taken together, MK could be an effective agent in reducing severity of ACS and associated complications. Experimental and clinical studies are warranted in this regard.

Author Contributions

Conceptualization, B.M.A. and H.M.A.-k.; methodology, B.M.A. and H.M.A.-k.; software, B.M.A. and H.M.A.-k.; validation, A.I.A.-G. and S.M.A.-H.; formal analysis, A.I.A.-G. and S.M.A.-H.; investigation, A.I.A.-G. and S.M.A.-H.; resources, A.I.A.-G. and S.M.A.-H.; data curation, A.I.A.-G. and S.M.A.-H.; writing—original draft preparation, H.M.A.-k., A.I.A.-G. and S.M.A.-H.; writing—review and editing, M.D.W., J.-M.S., H.M.S. and G.E.-S.B.; visualization, H.M.S.; supervision, G.E.-S.B.; project administration, G.E.-S.B.; funding acquisition, M.D.W. and J.-M.S. All authors have read and agreed to the published version of the manuscript.

Funding

M.D.W. thanks the French Agence Nationale de la Recherche and the Région Pays de la Loire for financial support on COVID-19 research (ANR Flash COVID 19 call—name: CoV2-E-TARGET—grant number: 2020 07132).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Al-Buhadilly, A.K. Rosuvastatin improves vaspin serum levels in obese patients with acute coronary syndrome. Diseases 2018, 6, 9. [Google Scholar] [CrossRef] [PubMed]
  2. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Samy, O.M. Statin therapy improves serum Annexin A1 levels in patients with acute coronary syndrome: A case-controlled study. Int. J. Crit. Illn. Inj. Sci. 2021, 11, 4. [Google Scholar] [CrossRef] [PubMed]
  3. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Awad, M.S.; Alrifai, S.B. Assessment of serum prolactin levels in acute myocardial infarction: The role of pharmacotherapy. Indian J. Endocrinol. Metab. 2016, 20, 72. [Google Scholar] [CrossRef] [PubMed]
  4. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Welson, N.N.; Batiha, G.E.-S. Trimetazidine and COVID-19-induced acute cardiac injury: A missed key. Int. J. Clin. Pharm. 2022, 44, 832–833. [Google Scholar] [CrossRef] [PubMed]
  5. Sami, O.M.; Al-kuraishy, H.M.; Al-Gareeb, A.I. Differential effects of statins on annexin A1 serum level in patients with acute coronary syndrome: A pleiotropic update. Ann. Trop. Med. Public Health 2021, 24, 394–402. [Google Scholar] [CrossRef]
  6. Mason, P.J.; Shah, B.; Tamis-Holland, J.E.; Bittl, J.A.; Cohen, M.G.; Safirstein, J.; Drachman, D.E.; Valle, J.A.; Rhodes, D.; Gilchrist, I.C. An update on radial artery access and best practices for transradial coronary angiography and intervention in acute coronary syndrome: A scientific statement from the American Heart Association. Circ. Cardiovasc. Interv. 2018, 11, e000035. [Google Scholar] [CrossRef]
  7. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef]
  8. Hoxha, M.; Rovati, G.E.; Cavanillas, A.B. The leukotriene receptor antagonist montelukast and its possible role in the cardiovascular field. Eur. J. Clin. Pharmacol. 2017, 73, 799–809. [Google Scholar] [CrossRef]
  9. Kellaway, C.; Trethewie, E. The liberation of a slow-reacting smooth muscle-stimulating substance in anaphylaxis. Q. J. Exp. Physiol. Cogn. Med. Sci. Transl. Integr. 1940, 30, 121–145. [Google Scholar] [CrossRef]
  10. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Almulaiky, Y.Q.; Cruz-Martins, N.; Batiha, G.E.-S. Role of leukotriene pathway and montelukast in pulmonary and extrapulmonary manifestations of Covid-19: The enigmatic entity. Eur. J. Pharmacol. 2021, 904, 174196. [Google Scholar] [CrossRef]
  11. Bankova, L.G.; Dwyer, D.F.; Yoshimoto, E.; Ualiyeva, S.; McGinty, J.W.; Raff, H.; von Moltke, J.; Kanaoka, Y.; Frank Austen, K.; Barrett, N.A. The cysteinyl leukotriene 3 receptor regulates expansion of IL-25–producing airway brush cells leading to type 2 inflammation. Sci. Immunol. 2018, 3, eaat9453. [Google Scholar] [CrossRef] [PubMed]
  12. He, R.; Chen, Y.; Cai, Q. The role of the LTB4-BLT1 axis in health and disease. Pharmacol. Res. 2020, 158, 104857. [Google Scholar] [CrossRef] [PubMed]
  13. Alorabi, M.; Cavalu, S.; Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Mostafa-Hedeab, G.; Negm, W.A.; Youssef, A.; El-Kadem, A.H.; Saad, H.M.; Batiha, G.E.-S. Pentoxifylline and berberine mitigate diclofenac-induced acute nephrotoxicity in male rats via modulation of inflammation and oxidative stress. Biomed. Pharmacother. 2022, 152, 113225. [Google Scholar] [CrossRef] [PubMed]
  14. Hayashi, H.; Fukutomi, Y.; Mitsui, C.; Kajiwara, K.; Watai, K.; Kamide, Y.; Nakamura, Y.; Hamada, Y.; Tomita, Y.; Sekiya, K. Omalizumab for aspirin hypersensitivity and leukotriene overproduction in aspirin-exacerbated respiratory disease. A randomized controlled trial. Am. J. Respir. Crit. Care Med. 2020, 201, 1488–1498. [Google Scholar] [CrossRef] [PubMed]
  15. Batiha, G.E.-S.; Al-Gareeb, A.I.; Saad, H.M.; Al-Kuraishy, H.M. COVID-19 and corticosteroids: A narrative review. Inflammopharmacology 2022, 30, 1189–1205. [Google Scholar] [CrossRef]
  16. Möller, I.; Murali, R.; Müller, H.; Wiesner, T.; Jackett, L.A.; Scholz, S.L.; Cosgarea, I.; van de Nes, J.A.; Sucker, A.; Hillen, U. Activating cysteinyl leukotriene receptor 2 (CYSLTR2) mutations in blue nevi. Mod. Pathol. 2017, 30, 350–356. [Google Scholar] [CrossRef]
  17. Tantisira, K.G.; Drazen, J.M. Genetics and pharmacogenetics of the leukotriene pathway. J. Allergy Clin. Immunol. 2009, 124, 422–427. [Google Scholar] [CrossRef]
  18. Park, J.S.; Chang, H.S.; Park, C.-S.; Lee, J.-H.; Lee, Y.M.; Choi, J.H.; Park, H.-S.; Kim, L.H.; Park, B.L.; Choi, Y.H. Association analysis of cysteinyl-leukotriene receptor 2 (CYSLTR2) polymorphisms with aspirin intolerance in asthmatics. Pharmacogenet. Genom. 2005, 15, 483–492. [Google Scholar] [CrossRef]
  19. Mostafa-Hedeab, G.; Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Jeandet, P.; Saad, H.M.; Batiha, G.E.-S. A raising dawn of pentoxifylline in management of inflammatory disorders in Covid-19. Inflammopharmacology 2022, 30, 799–809. [Google Scholar] [CrossRef]
  20. Itadani, S.; Yashiro, K.; Aratani, Y.; Sekiguchi, T.; Kinoshita, A.; Moriguchi, H.; Ohta, N.; Takahashi, S.; Ishida, A.; Tajima, Y. Discovery of gemilukast (ONO-6950), a dual CysLT1 and CysLT2 antagonist as a therapeutic agent for asthma. J. Med. Chem. 2015, 58, 6093–6113. [Google Scholar] [CrossRef]
  21. Calderón-Zamora, L.; Canizalez-Román, A.; León-Sicairos, N.; Aguilera-Mendez, A.; Huang, F.; Hong, E.; Villafaña, S. Changes in expression of orphan receptors GPR99 and GPR107 during the development and establishment of hypertension in spontaneously hypertensive rats. J. Recept. Signal Transduct. 2021, 41, 558–565. [Google Scholar] [CrossRef] [PubMed]
  22. Lecca, D.; Raffaele, S.; Abbracchio, M.P.; Fumagalli, M. Regulation and signaling of the GPR17 receptor in oligodendroglial cells. Glia 2020, 68, 1957–1967. [Google Scholar] [CrossRef] [PubMed]
  23. De Luca, L.; Steg, P.G.; Bhatt, D.L.; Capodanno, D.; Angiolillo, D.J. Cangrelor: Clinical data, contemporary use, and future perspectives. J. Am. Heart Assoc. 2021, 10, e022125. [Google Scholar] [CrossRef] [PubMed]
  24. Mansour, A.; Bachelot-Loza, C.; Nesseler, N.; Gaussem, P.; Gouin-Thibault, I. P2Y12 inhibition beyond thrombosis: Effects on inflammation. Int. J. Mol. Sci. 2020, 21, 1391. [Google Scholar] [CrossRef]
  25. Kang, J.H.; Lim, H.; Lee, D.S.; Yim, M. Montelukast inhibits RANKL-induced osteoclast formation and bone loss via CysLTR1 and P2Y12. Mol. Med. Rep. 2018, 18, 2387–2398. [Google Scholar] [CrossRef]
  26. Göbel, T.; Diehl, O.; Heering, J.; Merk, D.; Angioni, C.; Wittmann, S.K.; Buscato, E.l.; Kottke, R.; Weizel, L.; Schader, T. Zafirlukast is a dual modulator of human soluble epoxide hydrolase and peroxisome proliferator-activated receptor γ. Front. Pharmacol. 2019, 10, 263. [Google Scholar] [CrossRef]
  27. Fuse809 (talk)—Own work, Public Domain. Available online: https://commons.wikimedia.org/w/index.php?curid=31613539 (accessed on 18 August 2022).
  28. Sansing-Foster, V.; Haug, N.; Mosholder, A.; Cocoros, N.M.; Bradley, M.; Ma, Y.; Pennap, D.; Dee, E.C.; Toh, S.; Pestine, E. Risk of psychiatric adverse events among montelukast users. J. Allergy Clin. Immunol. Pract. 2021, 9, 385–393.e312. [Google Scholar] [CrossRef]
  29. Glockler-Lauf, S.D.; Finkelstein, Y.; Zhu, J.; Feldman, L.Y.; To, T. Montelukast and neuropsychiatric events in children with asthma: A nested case–control study. J. Pediatr. 2019, 209, 176–182.e174. [Google Scholar] [CrossRef]
  30. Fox, C.W.; Khaw, C.L.; Gerke, A.K.; Lund, B.C. Montelukast and neuropsychiatric events–a sequence symmetry analysis. J. Asthma 2021, 1–7. [Google Scholar] [CrossRef]
  31. Hirvensalo, P.; Tornio, A.; Neuvonen, M.; Tapaninen, T.; Paile-Hyvärinen, M.; Kärjä, V.; Männistö, V.T.; Pihlajamäki, J.; Backman, J.T.; Niemi, M. Comprehensive pharmacogenomic study reveals an important role of UGT1A3 in montelukast pharmacokinetics. Clin. Pharmacol. Ther. 2018, 104, 158–168. [Google Scholar] [CrossRef]
  32. Colazzo, F.; Gelosa, P.; Tremoli, E.; Sironi, L.; Castiglioni, L. Role of the cysteinyl leukotrienes in the pathogenesis and progression of cardiovascular diseases. Mediat. Inflamm. 2017, 2017, 2432958. [Google Scholar] [CrossRef] [PubMed]
  33. Folco, G.; Rossoni, G.; Buccellati, C.; Berti, F.; Maclouf, J.; Sala, A. Leukotrienes in cardiovascular diseases. Am. J. Respir. Crit. Care Med. 2000, 161, S112–S116. [Google Scholar] [CrossRef] [PubMed]
  34. Ibrahim, M.A.; Amin, E.F.; Ibrahim, S.A.; Abdelzaher, W.Y.; Abdelrahman, A.M. Montelukast and irbesartan ameliorate metabolic and hepatic disorders in fructose-induced metabolic syndrome in rats. Eur. J. Pharmacol. 2014, 724, 204–210. [Google Scholar] [CrossRef]
  35. Imig, J.D.; Hammock, B.D. Soluble epoxide hydrolase as a therapeutic target for cardiovascular diseases. Nat. Rev. Drug Discov. 2009, 8, 794–805. [Google Scholar] [CrossRef] [PubMed]
  36. Fleming, I. The pharmacology of the cytochrome P450 epoxygenase/soluble epoxide hydrolase axis in the vasculature and cardiovascular disease. Pharmacol. Rev. 2014, 66, 1106–1140. [Google Scholar] [CrossRef]
  37. Şener, G.; Şehirli, Ö.; Velioğlu-Öğünç, A.; Çetinel, Ş.; Gedik, N.; Caner, M.; Sakarcan, A.; Yeğen, B.Ç. Montelukast protects against renal ischemia/reperfusion injury in rats. Pharmacol. Res. 2006, 54, 65–71. [Google Scholar] [CrossRef] [PubMed]
  38. Ge, S.; Zhou, G.; Cheng, S.; Liu, D.; Xu, J.; Xu, G.; Liu, X. Anti-atherogenic effects of montelukast associated with reduced MCP-1 expression in a rabbit carotid balloon injury model. Atherosclerosis 2009, 205, 74–79. [Google Scholar] [CrossRef]
  39. Liu, D.; Ge, S.; Zhou, G.; Xu, G.; Zhang, R.; Zhu, W.; Liu, Z.; Cheng, S.; Liu, X. Montelukast inhibits matrix metalloproteinases expression in atherosclerotic rabbits. Cardiovasc. Drugs Ther. 2009, 23, 431–437. [Google Scholar] [CrossRef]
  40. Wang, Y.-X.J.; Ulu, A.; Zhang, L.-N.; Hammock, B. Soluble epoxide hydrolase in atherosclerosis. Curr. Atheroscler. Rep. 2010, 12, 174–183. [Google Scholar] [CrossRef]
  41. Cipollone, F.; Mezzetti, A.; Fazia, M.L.; Cuccurullo, C.; Iezzi, A.; Ucchino, S.; Spigonardo, F.; Bucci, M.; Cuccurullo, F.; Prescott, S.M. Association between 5-lipoxygenase expression and plaque instability in humans. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1665–1670. [Google Scholar] [CrossRef] [Green Version]
  42. Allayee, H.; Hartiala, J.; Lee, W.; Mehrabian, M.; Irvin, C.G.; Conti, D.V.; Lima, J.J. The effect of montelukast and low-dose theophylline on cardiovascular disease risk factors in asthmatics. Chest 2007, 132, 868–874. [Google Scholar] [CrossRef] [PubMed]
  43. Bäck, M. Leukotriene signaling in atherosclerosis and ischemia. Cardiovasc. Drugs Ther. 2009, 23, 41–48. [Google Scholar] [CrossRef] [PubMed]
  44. Helgadottir, A.; Manolescu, A.; Thorleifsson, G.; Gretarsdottir, S.; Jonsdottir, H.; Thorsteinsdottir, U.; Samani, N.J.; Gudmundsson, G.; Grant, S.F.; Thorgeirsson, G. The gene encoding 5-lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nat. Genet. 2004, 36, 233–239. [Google Scholar] [CrossRef] [PubMed]
  45. Mehrabian, M.; Allayee, H.; Wong, J.; Shih, W.; Wang, X.-P.; Shaposhnik, Z.; Funk, C.D.; Lusis, A.J. Identification of 5-lipoxygenase as a major gene contributing to atherosclerosis susceptibility in mice. Circ. Res. 2002, 91, 120–126. [Google Scholar] [CrossRef]
  46. Zhou, G.; Ge, S.; Liu, D.; Xu, G.; Zhang, R.; Yin, Q.; Zhu, W.; Chen, J.; Liu, X. Atorvastatin reduces plaque vulnerability in an atherosclerotic rabbit model by altering the 5-lipoxygenase pathway. Cardiology 2010, 115, 221–228. [Google Scholar] [CrossRef]
  47. Bevan, S.; Lorenz, M.W.; Sitzer, M.; Markus, H.S. Genetic variation in the leukotriene pathway and carotid intima-media thickness: A 2-stage replication study. Stroke 2009, 40, 696–701. [Google Scholar] [CrossRef]
  48. Dwyer, J.H.; Allayee, H.; Dwyer, K.M.; Fan, J.; Wu, H.; Mar, R.; Lusis, A.J.; Mehrabian, M. Arachidonate 5-lipoxygenase promoter genotype, dietary arachidonic acid, and atherosclerosis. N. Engl. J. Med. 2004, 350, 29–37. [Google Scholar] [CrossRef]
  49. Nobili, E.; Salvado, M.D.; Folkersen, L.; Castiglioni, L.; Kastrup, J.; Wetterholm, A.; Tremoli, E.; Hansson, G.K.; Sironi, L.; Haeggström, J.Z. Cysteinyl leukotriene signaling aggravates myocardial hypoxia in experimental atherosclerotic heart disease. PLoS ONE 2012, 7, e41786. [Google Scholar] [CrossRef]
  50. Duah, E.; Adapala, R.K.; Al-Azzam, N.; Kondeti, V.; Gombedza, F.; Thodeti, C.K.; Paruchuri, S. Cysteinyl leukotrienes regulate endothelial cell inflammatory and proliferative signals through CysLT2 and CysLT1 receptors. Sci. Rep. 2013, 3, 3274. [Google Scholar] [CrossRef]
  51. Cole, S.P. Multidrug resistance protein 1 (MRP1, ABCC1), a “multitasking” ATP-binding cassette (ABC) transporter. J. Biol. Chem. 2014, 289, 30880–30888. [Google Scholar] [CrossRef] [Green Version]
  52. Papayianni, A.; Serhan, C.N.; Brady, H.R. Lipoxin A4 and B4 inhibit leukotriene-stimulated interactions of human neutrophils and endothelial cells. J. Immunol. 1996, 156, 2264–2272. [Google Scholar] [PubMed]
  53. Uzonyi, B.; Lötzer, K.; Jahn, S.; Kramer, C.; Hildner, M.; Bretschneider, E.; Radke, D.; Beer, M.; Vollandt, R.; Evans, J.F. Cysteinyl leukotriene 2 receptor and protease-activated receptor 1 activate strongly correlated early genes in human endothelial cells. Proc. Natl. Acad. Sci. USA 2006, 103, 6326–6331. [Google Scholar] [CrossRef] [PubMed]
  54. Omede, A. Role of Alpha-Ketoglutarate Receptor G-Protein Coupled Receptor 99 (GPR99) in Cardiac Hypertrophy; The University of Manchester: Manchester, UK, 2015. [Google Scholar]
  55. Nguyen, A.V.; Bagood, M.D.; Wang, M.; Caryotakis, S.E.; Smith, G.; Yee, S.; Shen, H.; Isseroff, R.R.; Soulika, A.M. Montelukast, an Antagonist of Cysteinyl Leukotriene Signaling, Impairs Burn Wound Healing. Plast. Reconstr. Surg. 2022, 150, 92e–104e. [Google Scholar] [CrossRef]
  56. Wang, Y.; Li, C.; Liu, Z.; Shi, T.; Wang, Q.; Li, D.; Wu, Y.; Han, J.; Guo, S.; Tang, B.; et al. DanQi Pill protects against heart failure through the arachidonic acid metabolism pathway by attenuating different cyclooxygenases and leukotrienes B4. BMC Complement. Altern. Med. 2014, 14, 67. [Google Scholar] [CrossRef]
  57. Zhao, X.; Du, J.-Q.; Xu, D.-Y.; Zhao, S.-P. Effects of soluble epoxide hydrolase inhibitor on the expression of fatty acid synthase in peripheral blood mononuclear cell in patients with acute coronary syndrome. Lipids Heal. Dis. 2013, 12, 3. [Google Scholar] [CrossRef]
  58. Oni-Orisan, A.; Cresci, S.; Jones, P.G.; Theken, K.N.; Spertus, J.A.; Lee, C.R. Association between the EPHX2 p. Lys55Arg polymorphism and prognosis following an acute coronary syndrome. Prostaglandins Other Lipid Mediat. 2018, 138, 15–22. [Google Scholar] [CrossRef] [PubMed]
  59. Motoki, A.; Merkel, M.J.; Packwood, W.H.; Cao, Z.; Liu, L.; Iliff, J.; Alkayed, N.J.; Van Winkle, D.M. Soluble epoxide hydrolase inhibition and gene deletion are protective against myocardial ischemia-reperfusion injury in vivo. Am. J. Physiol.-Heart Circ. Physiol. 2008, 295, H2128–H2134. [Google Scholar] [CrossRef]
  60. Libby, P. Current concepts of the pathogenesis of the acute coronary syndromes. Circulation 2001, 104, 365–372. [Google Scholar] [CrossRef]
  61. Ali, S.A.-J.; Al-Shalah, H.; Al-Salihi, O. Assessment of circulating PPAR-γ Level as a risk and diagnostic biomarker in Acute Coronary Syndrome. J. Univ. Babylon Pure Appl. Sci. 2018, 26, 210–218. [Google Scholar] [CrossRef]
  62. Arat, A.; Yılmaz, Ü.; Yılmaz, N.; Fazlıoğulları, O.; Çelik, F.; Başaran, C.; Zeybek, Ü. Effects of Leptin, Resistin, and PPAR-Gama Gene Variants on Obese Patients with Acute Coronary Syndrome in the Turkish Population. JAREM. J. Acad. Res. Med. 2020, 10, 166. [Google Scholar] [CrossRef]
  63. Fujimori, K.; Uno, S.; Kuroda, K.; Matsumoto, C.; Maehara, T. Leukotriene C4 synthase is a novel PPARγ target gene, and leukotriene C4 and D4 activate adipogenesis through cysteinyl LT1 receptors in adipocytes. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2022, 1869, 119203. [Google Scholar] [CrossRef] [PubMed]
  64. Iovannisci, D.M.; Lammer, E.J.; Steiner, L.; Cheng, S.; Mahoney, L.T.; Davis, P.H.; Lauer, R.M.; Burns, T.L. Association between a leukotriene C4 synthase gene promoter polymorphism and coronary artery calcium in young women: The Muscatine Study. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 394–399. [Google Scholar] [CrossRef] [PubMed]
  65. Piomelli, D.; Feinmark, S.J.; Cannon, P.J. Leukotriene biosynthesis by canine and human coronary arteries. J. Pharmacol. Exp. Ther. 1987, 241, 763–770. [Google Scholar] [PubMed]
  66. Vigorito, C.; Giordano, A.; Cirillo, R.; Genovese, A.; Rengo, F.; Marone, G. Metabolic and hemodynamic effects of peptide leukotriene C4 and D4 in man. Int. J. Clin. Lab. Res. 1997, 27, 178–184. [Google Scholar] [CrossRef] [PubMed]
  67. Eaton, A.; Nagy, E.; Pacault, M.; Fauconnier, J.; Bäck, M. Cysteinyl leukotriene signaling through perinuclear CysLT1 receptors on vascular smooth muscle cells transduces nuclear calcium signaling and alterations of gene expression. J. Mol. Med. 2012, 90, 1223–1231. [Google Scholar] [CrossRef]
  68. Loötzer, K.; Spanbroek, R.; Hildner, M.; Urbach, A.; Heller, R.; Bretschneider, E.; Galczenski, H.; Evans, J.F.; Habenicht, A.J. Differential Leukotriene Receptor Expression and Calcium Responses in Endothelial Cells and Macrophages Indicate 5-Lipoxygenase–Dependent Circuits of Inflammation and Atherogenesis. Arterioscler. Thromb. Vasc. Biol. 2003, 23, e32–e36. [Google Scholar]
  69. Peters-Golden, M.; Henderson, W.R., Jr. Leukotrienes. N. Engl. J. Med. 2007, 357, 1841–1854. [Google Scholar] [CrossRef]
  70. Allen, S.P.; Sampson, A.P.; Piper, P.J.; Chester, A.H.; Ohri, S.K.; Yacoub, M.H. Enhanced excretion of urinary leukotriene E4 in coronary artery disease and after coronary artery bypass surgery. Coron. Artery Dis. 1993, 4, 899–904. [Google Scholar] [CrossRef]
  71. Ni, N.C.; Ballantyne, L.L.; Mewburn, J.D.; Funk, C.D. Multiple-site activation of the cysteinyl leukotriene receptor 2 is required for exacerbation of ischemia/reperfusion injury. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 321–330. [Google Scholar] [CrossRef]
  72. Gross, G.J.; Falck, J.R.; Gross, E.R.; Isbell, M.; Moore, J.; Nithipatikom, K. Cytochrome P450 and arachidonic acid metabolites: Role in myocardial ischemia/reperfusion injury revisited. Cardiovasc. Res. 2005, 68, 18–25. [Google Scholar] [CrossRef]
  73. Scott, W.; Pawlowski, N.; Andreach, M.; Cohn, Z. Resting macrophages produce distinct metabolites from exogenous arachidonic acid. J. Exp. Med. 1982, 155, 535–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Becher, U.M.; Ghanem, A.; Tiyerili, V.; Fürst, D.O.; Nickenig, G.; Mueller, C.F. Inhibition of leukotriene C4 action reduces oxidative stress and apoptosis in cardiomyocytes and impedes remodeling after myocardial injury. J. Mol. Cell. Cardiol. 2011, 50, 570–577. [Google Scholar] [CrossRef] [PubMed]
  75. Han, H.; Tian, R.; Neubauer, S.; Gaudron, P.; Hu, K.; Ertl, G. Effects of LTD4 and its specific antagonist L-660,711 in isolated rat hearts with chronic myocardial infarction. Am. J. Physiol.-Heart Circ. Physiol. 1994, 266, H2068–H2073. [Google Scholar] [CrossRef]
  76. Ni, N.C.; Yan, D.; Ballantyne, L.L.; Barajas-Espinosa, A.; Amand, T.S.; Pratt, D.A.; Funk, C.D. A selective cysteinyl leukotriene receptor 2 antagonist blocks myocardial ischemia/reperfusion injury and vascular permeability in mice. J. Pharmacol. Exp. Ther. 2011, 339, 768–778. [Google Scholar] [CrossRef] [PubMed]
  77. Jiang, W.; Hall, S.R.; Moos, M.P.; Cao, R.Y.; Ishii, S.; Ogunyankin, K.O.; Melo, L.G.; Funk, C.D. Endothelial cysteinyl leukotriene 2 receptor expression mediates myocardial ischemia-reperfusion injury. Am. J. Pathol. 2008, 172, 592–602. [Google Scholar] [CrossRef]
  78. Qi, A.-D.; Harden, T.K.; Nicholas, R.A. Is GPR17 a P2Y/leukotriene receptor? examination of uracil nucleotides, nucleotide sugars, and cysteinyl leukotrienes as agonists of GPR17. J. Pharmacol. Exp. Ther. 2013, 347, 38–46. [Google Scholar] [CrossRef]
  79. Carry, M.; Korley, V.; Willerson, J.T.; Weigelt, L.; Ford-Hutchinson, A.W.; Tagari, P. Increased urinary leukotriene excretion in patients with cardiac ischemia. In vivo evidence for 5-lipoxygenase activation. Circulation 1992, 85, 230–236. [Google Scholar] [CrossRef]
  80. Prescott, E.; Pernow, J.; Saraste, A.; Åkerblom, A.; Angerås, O.; Erlinge, D.; Grove, E.L.; Hedman, M.; Jensen, L.O.; Svedlund, S. Design and rationale of FLAVOUR: A phase IIa efficacy study of the 5-lipoxygenase activating protein antagonist AZD5718 in patients with recent myocardial infarction. Contemp. Clin. Trials Commun. 2020, 19, 100629. [Google Scholar] [CrossRef]
  81. Lee, C.C.; Appleyard, R.F.; Byrne, J.G.; Cohn, L.H. Leukotrienes D4 and E4 produced in myocardium impair coronary flow and ventricular function after two hours of global ischaemia in rat heart. Cardiovasc. Res. 1993, 27, 770–773. [Google Scholar] [CrossRef]
  82. Rossoni, G.; Sala, A.; Berti, F.; Testa, T.; Buccellati, C.; Molta, C.; Muller-Peddinghaus, R.; Maclouf, J.; Folco, G. Myocardial protection by the leukotriene synthesis inhibitor BAY X1005: Importance of transcellular biosynthesis of cysteinyl-leukotrienes. J. Pharmacol. Exp. Ther. 1996, 276, 335–341. [Google Scholar]
  83. Shekher, A.; Singh, M. Role of eicosanoid inhibition of ischemia reperfusion injury: Intact and isolated rat heart studies. Methods Find. Exp. Clin. Pharmacol. 1997, 19, 223–229. [Google Scholar] [PubMed]
  84. Hahn, R.A.; MacDonald, B.R.; Simpson, P.J.; Potts, B.D.; Parli, C.J. Antagonism of leukotriene B4 receptors does not limit canine myocardial infarct size. J. Pharmacol. Exp. Ther. 1990, 253, 58–66. [Google Scholar] [PubMed]
  85. Helgadottir, A.; Manolescu, A.; Helgason, A.; Thorleifsson, G.; Thorsteinsdottir, U.; Gudbjartsson, D.F.; Gretarsdottir, S.; Magnusson, K.P.; Gudmundsson, G.; Hicks, A. A variant of the gene encoding leukotriene A4 hydrolase confers ethnicity-specific risk of myocardial infarction. Nat. Genet. 2006, 38, 68–74. [Google Scholar] [CrossRef] [PubMed]
  86. Horii, Y.; Nakaya, M.; Ohara, H.; Nishihara, H.; Watari, K.; Nagasaka, A.; Nakaya, T.; Sugiura, Y.; Okuno, T.; Koga, T. Leukotriene B4 receptor 1 exacerbates inflammation following myocardial infarction. FASEB J. 2020, 34, 8749–8763. [Google Scholar] [CrossRef] [PubMed]
  87. Hoxha, M.; Lewis-Mikhael, A.-M.; Bueno-Cavanillas, A. Potential role of leukotriene receptor antagonists in reducing cardiovascular and cerbrovascular risk: A systematic review of human clinical trials and in vivo animal studies. Biomed. Pharmacother. 2018, 106, 956–965. [Google Scholar] [CrossRef]
  88. Khodir, A.; Ghoneim, H.; Rahim, M.; Suddek, G. Montelukast attenuates lipopolysaccharide-induced cardiac injury in rats. Hum. Exp. Toxicol. 2016, 35, 388–397. [Google Scholar] [CrossRef]
  89. Stodółkiewicz, E.; Sokołowska, B.; Rzeszutko, M.; Tomala, M.; Chrustowicz, A.; Żmudka, K.; Sanak, M.; Szczeklik, W. Leukotriene biosynthesis in coronary artery disease: Results of the Leukotrienes and Thromboxane In Myocardial Infarction (LTIMI) study. Pol. Arch. Med. Wewnętrznej Pol. Arch. Intern. Med. 2018, 128, 43–51. [Google Scholar] [CrossRef]
  90. Vroegindewey, M.M.; Buljubasic, N.; Oemrawsingh, R.M.; Kardys, I.; Asselbergs, F.W.; van der Harst, P.; Umans, V.A.; Kietselaer, B.; Lenderink, T.; Liem, A. MAPK-cascade stimulating biomarkers in relation to recurrent coronary events following an acute coronary syndrome. In The Prognostic Value of Coronary Imaging and Biomarkers in Ischemic Heart Disease; Gildeprint: Enschede, The Netherlands, 2019; p. 123. [Google Scholar]
  91. Wang, L.; Li, D.; Yang, K.; Hu, Y.; Zeng, Q. Toll-like receptor-4 and mitogen-activated protein kinase signal system are involved in activation of dendritic cells in patients with acute coronary syndrome. Immunology 2008, 125, 122–130. [Google Scholar] [CrossRef]
  92. Indolfi, C.; Gasparri, C.; Vicinanza, C.; De Serio, D.; Boncompagni, D.; Mongiardo, A.; Spaccarotella, C.; Agosti, V.; Torella, D.; Curcio, A. Mitogen-activated protein kinases activation in T lymphocytes of patients with acute coronary syndromes. Basic Res. Cardiol. 2011, 106, 667–679. [Google Scholar] [CrossRef]
  93. Wang, S.; Su, X.; Xu, L.; Chang, C.; Yao, Y.; Komal, S.; Cha, X.; Zang, M.; Ouyang, X.; Zhang, L. Glycogen synthase kinase-3β inhibition alleviates activation of the NLRP3 inflammasome in myocardial infarction. J. Mol. Cell. Cardiol. 2020, 149, 82–94. [Google Scholar] [CrossRef]
  94. Mauro, A.G.; Bonaventura, A.; Mezzaroma, E.; Quader, M.; Toldo, S. NLRP3 inflammasome in acute myocardial infarction. J. Cardiovasc. Pharmacol. 2019, 74, 175–187. [Google Scholar] [CrossRef] [PubMed]
  95. Yin, Y.; Zhou, Z.; Liu, W.; Chang, Q.; Sun, G.; Dai, Y. Vascular endothelial cells senescence is associated with NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation via reactive oxygen species (ROS)/thioredoxin-interacting protein (TXNIP) pathway. Int. J. Biochem. Cell Biol. 2017, 84, 22–34. [Google Scholar] [CrossRef] [PubMed]
  96. Suhara, T.; Baba, Y.; Shimada, B.K.; Higa, J.K.; Matsui, T. The mTOR signaling pathway in myocardial dysfunction in type 2 diabetes mellitus. Curr. Diab. Rep. 2017, 17, 38. [Google Scholar] [CrossRef]
  97. Blackburn, N.J.R.; Vulesevic, B.; McNeill, B.; Cimenci, C.E.; Ahmadi, A.; Gonzalez-Gomez, M.; Ostojic, A.; Zhong, Z.; Brownlee, M.; Beisswenger, P.J.; et al. Methylglyoxal-derived advanced glycation end products contribute to negative cardiac remodeling and dysfunction post-myocardial infarction. Basic Res. Cardiol. 2017, 112, 57. [Google Scholar] [CrossRef] [PubMed]
  98. Mansour, R.M.; Ahmed, M.A.; El-Sahar, A.E.; El Sayed, N.S. Montelukast attenuates rotenone-induced microglial activation/p38 MAPK expression in rats: Possible role of its antioxidant, anti-inflammatory and antiapoptotic effects. Toxicol. Appl. Pharmacol. 2018, 358, 76–85. [Google Scholar] [CrossRef] [PubMed]
  99. Zhou, C.; Shi, X.; Huang, H.; Zhu, Y.; Wu, Y. Montelukast attenuates neuropathic pain through inhibiting p38 mitogen-activated protein kinase and nuclear factor-kappa B in a rat model of chronic constriction injury. Anesth. Analg. 2014, 118, 1090–1096. [Google Scholar] [CrossRef]
  100. Fei, Z.; Zhang, L.; Wang, L.; Jiang, H.; Peng, A. Montelukast ameliorated pemetrexed-induced cytotoxicity in hepatocytes by mitigating endoplasmic reticulum (ER) stress and nucleotide oligomerization domain-like receptor protein 3 (NLRP3) activation. Bioengineered 2022, 13, 7894–7903. [Google Scholar] [CrossRef]
  101. El-Kashef, D.H.; Zaghloul, R.A. Ameliorative effect of montelukast against carbon tetrachloride-induced hepatotoxicity: Targeting NLRP3 inflammasome pathway. Life Sci. 2022, 304, 120707. [Google Scholar] [CrossRef]
  102. Tong, J.; Yu, Q.; Xu, W.; Yu, W.; Wu, C.; Wu, Y.; Yan, H. Montelukast enhances cytocidal effects of carfilzomib in multiple myeloma by inhibiting mTOR pathway. Cancer Biol. Ther. 2019, 20, 381–390. [Google Scholar] [CrossRef] [PubMed]
  103. Lei, C.; Wu, S.; Wen, C.; Li, Y.; Liu, N.; Huang, J.; Li, L.; Fu, M.; Liu, J. Zafirlukast attenuates advanced glycation end-products (AGEs)-induced degradation of articular extracellular matrix (ECM). Int. Immunopharmacol. 2019, 68, 68–73. [Google Scholar] [CrossRef]
  104. Abbate, R.; Cioni, G.; Ricci, I.; Miranda, M.; Gori, A.M. Thrombosis and acute coronary syndrome. Thromb. Res. 2012, 129, 235–240. [Google Scholar] [CrossRef] [PubMed]
  105. Carter, A.M. Inflammation, thrombosis and acute coronary syndromes. Diabetes Vasc. Dis. Res. 2005, 2, 113–121. [Google Scholar] [CrossRef]
  106. Duman, H.; Çinier, G.; Bakırcı, E.M.; Duman, H.; Şimşek, Z.; Hamur, H.; Değirmenci, H.; Emlek, N. Relationship between C-reactive protein to albumin ratio and thrombus burden in patients with acute coronary syndrome. Clin. Appl. Thromb. Hemost. 2019, 25, 1076029618824418. [Google Scholar] [CrossRef] [PubMed]
  107. Gorog, D.A.; Price, S.; Sibbing, D.; Baumbach, A.; Capodanno, D.; Gigante, B.; Halvorsen, S.; Huber, K.; Lettino, M.; Leonardi, S.; et al. Antithrombotic therapy in patients with acute coronary syndrome complicated by cardiogenic shock or out-of-hospital cardiac arrest: A joint position paper from the European Society of Cardiology (ESC) Working Group on Thrombosis, in association with the Acute Cardiovascular Care Association (ACCA) and European Association of Percutaneous Cardiovascular Interventions (EAPCI). Eur. Heart J. Cardiovasc. Pharmacother. 2021, 7, 125–140. [Google Scholar] [CrossRef] [PubMed]
  108. Babalghith, A.O.; Al-kuraishy, H.M.; Al-Gareeb, A.I.; De Waard, M.; Sabatier, J.-M.; Saad, H.M.; Batiha, G.E.-S. The Potential Role of Growth Differentiation Factor 15 in COVID-19: A Corollary Subjective Effect or Not? Diagnostics 2022, 12, 2051. [Google Scholar] [CrossRef]
  109. Wang, D.; Zhou, Y.; Qi, Y.; Song, M.; Yao, H.; Liao, C.; Lin, H.; Huang, M.; Zhuo, D.; Jiang, L. Antithrombotic Effects of Montelukast by Targeting Coagulation Factor XIa. Preprint. 2021. [Google Scholar]
  110. Houard, X.; Ollivier, V.; Louedec, L.; Michel, J.B.; Back, M. Differential inflammatory activity across human abdominal aortic aneurysms reveals neutrophilderived leukotriene B4 as a major chemotactic factor released from the intraluminal thrombus. FASEB J. 2009, 23, 1376–1383. [Google Scholar] [CrossRef]
  111. Datta, Y.H.; Romano, M.; Jacobson, B.C.; Golan, D.E.; Serhan, C.N.; Ewenstein, B.M. Peptido-leukotrienes are potent agonists of von Willebrand factor secretion and P-selectin surface expression in human umbilical vein endothelial cells. Circulation 1995, 92, 3304–3311. [Google Scholar] [CrossRef]
  112. Chan, S.J.; Ng, M.P.; Zhao, H.; Ng, G.J.; De Foo, C.; Wong, P.T.-H.; Seet, R. Early and sustained increases in leukotriene B4 levels are associated with poor clinical outcome in ischemic stroke patients. Neurotherapeutics 2020, 17, 282–293. [Google Scholar] [CrossRef]
  113. Nichols, W.W.; Mehta, J.L.; Thompson, L.; Donnelly, W.H. Synergistic effects of LTC4 and TxA2 on coronary flow and myocardial function. Am. J. Physiol.-Heart Circ. Physiol. 1988, 255, H153–H159. [Google Scholar] [CrossRef]
  114. Al-kuraishy, H.M.; Batiha, G.E.-S.; Faidah, H.; Al-Gareeb, A.I.; Saad, H.M.; Simal-Gandara, J. Pirfenidone and post-Covid-19 pulmonary fibrosis: Invoked again for realistic goals. Inflammopharmacology 2022, 1–10. [Google Scholar] [CrossRef]
  115. Tang, X.; Fuchs, D.; Tan, S.; Trauelsen, M.; Schwartz, T.W.; Wheelock, C.E.; Li, N.; Haeggström, J.Z. Activation of metabolite receptor GPR91 promotes platelet aggregation and transcellular biosynthesis of leukotriene C4. J. Thromb. Haemost. 2020, 18, 976–984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Leukotriene pathway and its inhibitors: Membrane phospholipids hydrolyzed by phospholipase A to arachidonic acid (AA), which, by the action of 5-lipoxygenase (5-LO), convert to leukotriene A4 (LTA4). Further, LTA4 is converted to LTB4 by the action of LTA4 hydrolase and to cysteinleukotriene (CysLT) by the action of LTC4 synthase. CysLTs act on CysLT receptors (CysLT1R and CysLT2R).
Figure 1. Leukotriene pathway and its inhibitors: Membrane phospholipids hydrolyzed by phospholipase A to arachidonic acid (AA), which, by the action of 5-lipoxygenase (5-LO), convert to leukotriene A4 (LTA4). Further, LTA4 is converted to LTB4 by the action of LTA4 hydrolase and to cysteinleukotriene (CysLT) by the action of LTC4 synthase. CysLTs act on CysLT receptors (CysLT1R and CysLT2R).
Pharmaceuticals 15 01147 g001
Figure 2. Chemical structure of montelukast (Reprinted from Ref. [27]).
Figure 2. Chemical structure of montelukast (Reprinted from Ref. [27]).
Pharmaceuticals 15 01147 g002
Figure 3. Mechanisms of montelukast in different cardiac and vascular diseases: Montelukast (MK) inhibits inflammasome signaling pathways, including p38 mitogen-activated protein kinase (p38MAPK), nod-like receptor pyrin 3 (NLRP3) inflammasome, mechanistic target of rapamycin (mTOR), and advanced glycation end-products (AGEPs). In addition, MK attenuates activation of cysteinleukotrine (CysLT) and activated factorXI with subsequent inhibition of inflammation and thrombosis-induced myocardial infarction and acute coronary syndrome (ACS).
Figure 3. Mechanisms of montelukast in different cardiac and vascular diseases: Montelukast (MK) inhibits inflammasome signaling pathways, including p38 mitogen-activated protein kinase (p38MAPK), nod-like receptor pyrin 3 (NLRP3) inflammasome, mechanistic target of rapamycin (mTOR), and advanced glycation end-products (AGEPs). In addition, MK attenuates activation of cysteinleukotrine (CysLT) and activated factorXI with subsequent inhibition of inflammation and thrombosis-induced myocardial infarction and acute coronary syndrome (ACS).
Pharmaceuticals 15 01147 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alomair, B.M.; Al-kuraishy, H.M.; Al-Gareeb, A.I.; Al-Hamash, S.M.; De Waard, M.; Sabatier, J.-M.; Saad, H.M.; El-Saber Batiha, G. Montelukast and Acute Coronary Syndrome: The Endowed Drug. Pharmaceuticals 2022, 15, 1147. https://doi.org/10.3390/ph15091147

AMA Style

Alomair BM, Al-kuraishy HM, Al-Gareeb AI, Al-Hamash SM, De Waard M, Sabatier J-M, Saad HM, El-Saber Batiha G. Montelukast and Acute Coronary Syndrome: The Endowed Drug. Pharmaceuticals. 2022; 15(9):1147. https://doi.org/10.3390/ph15091147

Chicago/Turabian Style

Alomair, Basil Mohammed, Hayder M. Al-kuraishy, Ali I. Al-Gareeb, Sadiq M. Al-Hamash, Michel De Waard, Jean-Marc Sabatier, Hebatallah M. Saad, and Gaber El-Saber Batiha. 2022. "Montelukast and Acute Coronary Syndrome: The Endowed Drug" Pharmaceuticals 15, no. 9: 1147. https://doi.org/10.3390/ph15091147

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