Abstract
Nicorandil is a dual mechanism anti-anginal agent that acts as a nitric oxide (NO) donor and a potassium (K+) channel opener. Recent studies have evaluated the effect of nicorandil on ischemic stroke. Neurons have a low tolerance to hypoxia and therefore the brain tissue is significantly vulnerable to ischemia. Current approved treatments for ischemic stroke are tissue plasminogen activators and clot retrieval methods. The narrow therapeutic time window and lack of efficacy in restoring the dying neurons urge researchers to develop an alternative approach.
In the terminal stages of anoxia, K+ channels induce hyperpolarization in various types of neuronal cells, leading to decreased neuronal activity and the preservation of the brain's energy. Nicorandil can open these K+ channels and sustain the hyperpolarization phase, which may have a neuroprotective effect during hypoxia. Additionally, we review how nicorandil can improve overall stroke outcomes through its anti-inflammatory, anti-oxidative, and edema-reducing effects. One of the major components evaluated in stroke patients is blood pressure. Studies have demonstrated that the effect of nicorandil on blood pressure is related to both its K+ channel opening and NO donating mechanisms. Since both hypertension and hypotension need correction before stroke intervention, it's crucial to consider the role of nicorandil and its impact on blood pressure. Previously published studies indicate that the right dosage of nicorandil can improve cerebral blood flow without significant changes in hemodynamic profiles. In this review, we discuss how nicorandil may contribute to better stroke outcomes based on previously published literature and laboratory findings.
Keywords: Ischemic stroke, Nicorandil, K+ channels, Nitric-oxide, Blood pressure
Graphical abstract

Highlights
-
•
K+ channels can be found in neurons and contribute to neuroprotection.
-
•
KATP channels preserve the brain energy during ischemia.
-
•
Nicorandil contributes to neuroprotection through diverse mechanisms.
-
•
Nicorandil's effect on blood pressure is contributable to both its K+ channel opening and nitric oxide donating mechanism.
-
•
Nicorandil improves cerebral circulation without affecting hemodynamic profiles.
1. Introduction
Stroke is among the leading causes of morbidity and mortality worldwide [1]. It is a result of either sudden loss of blood perfusion to the brain known as ischemic stroke (IS) or a rupture of cerebral blood vessels causing intra-cerebral hemorrhage [2]. In an individual presenting with IS, there is an area in the brain that receives between 10% and 30% of its normal blood flow called the “penumbra” [3]. This tissue, unlike the ischemic core, has not undergone irreversible infarction yet, but the neurons are electrically malfunctioning and causing neurological deficits. The principle of treatment is to restore blood to the penumbra and prevent permanent neuron death.
To date, only tissue plasminogen activator (tPA) and mechanical thrombectomy are approved for restoring blood perfusion to the ischemic area [4,5]. Only a small percentage of individuals (maximum of 5%) are eligible to receive tPA therapy in the acute phase of IS, mainly due to delays in getting to the hospital or the narrow therapeutic time window [6]. The recanalization efficacy of intra-venous tPA is limited in IS [7]. Therefore, not only many patients do not meet the criteria for tPA therapy, but for a significant number of patients, the tPA therapy is ineffective. Mechanical thrombectomy is indicated for individuals experiencing acute ischemic stroke resulting from a significant blockage in a major artery within the brain's front region, provided that treatment can be initiated within 24 h from when their last known normal neurological state was observed. Two challenges may restrict the use of mechanical thrombectomy, Firstly, only around 10% of patients with acute ischemic stroke exhibit a substantial blockage in a major artery in the front part of the brain and present themselves for eligibility within a 6-h timeframe [[8], [9], [10]]. Secondly, only a limited number of stroke centers possess the necessary resources and specialized skills to provide this treatment [11]. That being said, due to the limitations in currently approved therapies for IS, it is crucial to investigate further treatments that could improve stroke outcomes for patients not eligible or who have failed these treatments.
The underlying mechanisms of ischemia for acute myocardial infarction (AMI) and acute IS are similar [12]. Nicorandil is a hybrid of an adenosine triphosphate (ATP)-sensitive potassium (K+) channel (KATP) opener and nitrates. It can increase the outflow of K+ from cells, inhibit the inflow of calcium (Ca2+), reduce the Ca2+ overload, and consequently reduce the incidence of arrhythmia. It possesses the potential to expand the small coronary arteries and increase the coronary blood flow. These effects could be beneficial in the acute phase of IS [13]. Furthermore, animal studies have shown that nicorandil has therapeutic effects on various nervous system disorders, including its ability to protect against spinal cord issues, cerebral ischemia, and brain injuries by diminishing oxidative damage, mitigating inflammatory reactions, and employing anti-apoptotic mechanisms [[14], [15], [16]].
2. Nicorandil
Nicorandil is a nicotinamide nitrate with vasodilatory and K+ channel opening effects.
Its cardioprotective effects are attributable to its impacts on KATP channels which protect the ischemic tissue from losing ATP [[17], [18], [19]]. When consumed, nicorandil is primarily metabolized through oxidation to nicorandil-N-oxide and hydroxy nicorandil, and by denitration to N-(2-hydroxyethyl)-nicotin-amide. It is then transformed into nicotinamide, nicotinic acid, and N-methyl-nicotinamide. It has a half-life of 45 min during the first rapid phase of elimination after oral or intravenous administration [20]. Nicorandil acts as both a KATP channel opener and a nitric oxide (NO) donor [21].
3. Nicorandil and ischemic stroke
Ischemic conditions deprive the affected organ of the oxygen and nutrients needed for normal function. Neurons have a low tolerance to severe hypoxia resulting in cerebral anoxia [22]. The absence of oxygen and the resulting anoxia lead to the extracellular accumulation of glutamate, which can be excitotoxic [23]. It appears that anoxia-induced vulnerability can be related to an accompanied rise in intracellular Ca2+ concentration caused by increased glutamate concentrations in the interstitial space. Ca2+ elevations in such neurons are primarily attributed to influx through Ca2+-permeable glutamate receptors and voltage-activated Ca2+ channels during anaerobic depolarization [22,24,25]. Furthermore, inhibition of ATP-dependent ion pumps, such as the Na+/K+-ATPase, impairs Ca2+ extrusion and therefore, contributes to an excessive rise in Ca2+ [25]. The Ca2+ overload and ATP consumption by depolarized mitochondria lead to a cascade of molecular pathologies that ultimately result in cell death [23,26].
In a variety of neurons, a K+ channel induces hyperpolarization following the terminal anoxic depolarization [22,27]. This hyperpolarization leads to decreased neuronal activity and therefore, transmembrane ion fluxes which diminish the activity of ion pumps that absorb 50% of the brain's energy [28]. That being said, if the K+ channels persist in the hyperpolarization phase for a reasonable amount of time, it might have a protective effect during brain hypoxia [29]. It has been demonstrated that KATP channels mediate hypoxic hyperpolarization [30,31]. KATP channels are subtypes of K+ channels that conduct inward-rectifier potassium current in response to cellular energy depletion. They are members of the inwardly rectifying K+ (Kir) superfamily and are multimers of four pore-forming Kir6 subunits and four Sulfonyl-Urea Receptor (SUR) subunits [32]. KATP channels are present in both the cellular membrane and the inner mitochondrial membrane, and they have been identified in various tissues, including the kidney, pancreatic beta cells, muscles, heart, and the nervous system [32].
3.1. Cardiovascular KATP channels and protection from ischemic injury
Nicorandil attends effective therapy in AMI patients which has resulted in reducing the incidence of reperfusion injury and myocardial infarct size [33]. The opening of mitochondrial KATP channels leads to increased K+ entry into the mitochondria and inhibits the depolarization of the cell membrane, thereby preventing mitochondrial Ca2+ overload and protecting the heart [28]. Nicorandil also leads to vasodilation as a result of vascular smooth muscle cell hyperpolarization [34] that confers its vasodilator activity [28], causing a reduction in systemic vascular resistance, mean arterial pressure, pulmonary capillary wedge pressure, and left ventricular end-diastolic pressure. Additionally, nicorandil dilates large coronary arteries, decreases coronary vascular resistance, and increases coronary artery blood flow. According to a meta-analysis, treatment with nicorandil as an adjunct to reperfusion and percutaneous coronary intervention therapy improves heart functional outcomes after AMI by improving the microvascular function, reducing infarct size, and increasing the left ventricular ejection fraction [35]. This study demonstrated that nicorandil decreased all-cause mortality in coronary artery disease patients irrespective of the delivery route of this drug [35].
3.2. Cerebral KATP channels and possible protection from ischemic injury
In the brain, KATP channels are present in the substantia nigra, neocortex, hippocampus, and hypothalamus. These channels are highly expressed in interneurons and granule cells, which are resistant to ischemic injury [32]. A previous study has demonstrated that brain mitochondria contain six to seven times more mitochondrial KATP channels compared with the mitochondria in the heart tissue [36]. The expression of subunits differs across the brain. Neurons mostly express Kir6.2 and SUR1, whereas astrocytes express Kir6.1, SUR1, and SUR2 subunits [32].
The neuroprotective role of mitochondrial KATP channels in cerebral ischemia has been investigated [32]. It has been revealed that cortical neurons in Kir6.2 (−/−) KATP channel knockout mice are more vulnerable to ischemia compared with the wild-type neurons in a middle cerebral artery occlusion (MCAO) model [32]. Also, overexpression of the Kir6.2 leads to high resistance to ischemia and a reduction in ischemic brain damage in cortical neurons [37]. The tolerance to anoxia may be due to a long-term hyperpolarization mediated by KATP channels [29]. KATP channel openers have been shown to attenuate cortical neuronal death induced by oxygen-glucose deprivation (OGD) in rats [38]. Pre-conditioning of neural stem cells (NSC) using nicorandil has shown increased brain-derived neurotrophic factor (BDNF). BDNF is a neurotrophic factor that modifies neurogenesis and survival signaling pathways. It has also been observed that BDNF expression in preconditioned NSC led to activation of the phosphoinositide 3-kinases (PI3K) activity. PI3K is an enzyme involved in cellular functions that can lead to cell survival and simulate neurite growth [39,40]. This study also demonstrated the anti-inflammatory properties of nicorandil-preconditioned NSC, where the preconditioning could downregulate pro-inflammatory cytokines (IL-1β, IL-6, IL-12, and TNF-α) and increase the production of anti-inflammatory mediators (IL-10) [41].
Due to the restricted oxygen and nutrient delivery and the consequent decrease in the ATP/ADP ratio that happens during cerebral ischemia, KATP channels open and cause neuronal hyperpolarization. A decrease in the ionic pump activity reduces the metabolic needs of the neurons and protects them against excitotoxicity that occurs during anoxia [29,32]. Few in vivo and in vitro studies evaluated the efficacy of nicorandil as a KATP channel opener in treating stroke [14,15].
Nicorandil has the potential to mitigate the progression of cerebral ischemia by alleviating factors such as Ca2+ overload, brain edema, and oxidative stress, and by demonstrating anti-inflammatory and anti-apoptotic properties. We describe the possible treatment effects of nicorandil on IS.
3.3. Role of nicorandil in cerebral ischemia
3.3.1. Reducing calcium overload
Nicorandil can protect against ischemic injury through diverse mechanisms. As described earlier, Ca2+ rise is one of the major causes of cell death. The dysfunction of the Na+/k+ ATPase pump as a result of ischemia occurs within minutes and leads to the accumulation of Ca2+ and activation of Ca2+-dependent enzymes [42]. The opening of mitochondrial KATP channels decreases mitochondrial Ca2+ overload. A decrease in mitochondrial Ca2+ inhibits the opening of the mitochondrial permeability transition pore, a transmembrane protein residing in the mitochondrial inner membrane. This pore opens during reperfusion and is responsible for mitochondrial swelling and initiation of mitochondria-dependent apoptosis [43]. Activation of the KATP channel can also decrease intracellular Ca2+ levels. Nicorandil has been shown to alleviate intracellular Ca2+ overload in a rat cardiomyocyte cell line when the cells underwent hypoxia/reoxygenation injury [44]. Another study also showed that nicorandil reduced intracellular Ca2+ accumulation induced by OGD in the rat hippocampal CA1 pyramidal cells [45].
3.3.2. Reducing brain edema
IS leads to cerebral edema due to the disruption of the blood-brain barrier (BBB) during the first 24–48 h, and reaches a maximum extent of 3–5 days from the occurrence of IS [42]. The BBB disruption improperly permits serum proteins to shift from the blood to the brain parenchyma and lead to a type of vasogenic edema [46]. Ca2+ influx into the cells plays a key role in damaging the BBB [47]. KATP channel openers such as nicorandil can decrease intracellular Ca2+ which can help preserve the BBB integrity [45]. It has been observed that aquaporins 4 (AQP-4) (plasma membrane channels specialized for water transport) are involved in ischemic brain damage and brain edema [48]. The effect of nicorandil on cerebral injury following cardiac arrest was investigated and showed that AQP-4 expression in the brain tissue of pigs was significantly lower in the nicorandil-treated group than in the control group 6 h after the return of systemic circulation. The decreased expression of AQP-4 induced by nicorandil might translate into a reduction in the degree of brain edema after ischemia [49]. In a rat MCAO model, nicorandil administration for 3 days reduced cerebral edema, which was determined by measuring the brain water content [14].
3.3.3. Anti-oxidative effect
Ischemia induces the production of reactive oxygen species that can damage cellular components [42]. In an in vitro study on astrocytes, nicorandil treatment decreased the expression of endoplasmic reticulum stress-related proteins in response to OGD [50]. Nicorandil has demonstrated neuroprotective effects against H2O2-induced neuronal stress [51] and inhibited the production of reactive oxygen species in endothelial cells under oxidative stress [52]. In a mouse model of 2-vessel occlusion, nicorandil administration has shown a decrease in oxidative stress markers such as catalase and superoxide dismutase in the brain [53].
3.3.4. Reducing inflammation
Increased infiltration of peripheral leukocytes into the brain has been observed hours to days after ischemic injury [42]. Activated microglia, which are the brain's resident inflammatory cells, become phagocytic and migrate to the ischemic core. This triggers an immune response and release of interleukins such as IL-1 β, tumor necrosis factor (TNF), and IL-6 [54]. The pro-inflammatory cytokines that have been activated after IS disrupt the BBB which results in further infiltration of inflammatory cells [55,56]. Release of the chemokine matrix metalloproteinases (MMP), particularly MMP-9, by the innate immune system results in further disruption of the BBB leading to edema and growth of the infarct area [57] Downregulation of immune components has been found to contribute to better stroke outcomes [58]. KATP channels are likely involved in regulating the production of IL-1β by microglia after OGD. The pre-treatment of the microglial cell line with nicorandil can decrease the production of IL-1β [59]. Treatment with nicorandil has been shown to attenuate the inflammatory response induced by OGD [50] and decrease inflammatory markers in the brain after cerebral ischemia [53]. Nicorandil has also been shown to inhibit platelet activation, probably by increasing the activity of endothelial NO [52]. In a swine cardiac arrest model evaluating the effect of nicorandil on the resulting cerebral injury, TNF-α and IL-6 levels were significantly lower in the nicorandil group than in the control group after three and 6 h of the return of systemic circulation [16]. In a clinical study on 157 patients with unstable angina who received nicorandil in addition to the conventional treatment, high-sensitivity C-reactive protein and MMP-9 were significantly decreased 21 days after nicorandil treatment compared with the control group, who merely received the conventional treatment [60].
3.3.5. Anti-apoptosis
Apoptosis within the ischemic penumbra may occur after a substantial time delay [42]. In an in vitro study, nicorandil has been shown to protect cultured glioblastoma cells against H2O2-induced cell death [51]. Nicorandil also inhibited apoptosis induced by oxidative stress as evidenced by the suppression of the activity of caspase-3 in cerebellar granule neurons [61] and a neuroblastoma cell line in a cellular model of Alzheimer's disease [62]. Another study showed nicorandil could cause more neuronal density and less neuronal death in the face of ischemia [63] and that the effect of nicorandil was dose-dependent, as mice treated with higher doses of nicorandil (10 mg/kg and 20 mg/kg) showed a more considerable reduction in the number of TUNEL-positive cells than those treated with a low dose (5 mg/kg) [63]. They concluded that nicorandil has a neuroprotective role in ischemia-reperfusion (I/R) injury induced by deep hypothermic low flow by inhibiting apoptosis through the activation of the PI3K/Akt1 signaling pathway [63].
4. Effects of nicorandil in animal studies
The oral administration of 2 and 4 mg/kg of nicorandil once a day from the 7th day post-stroke until the 30th day caused a significant decrease in the infarct size and serum nitrosative stress, brain oxidative stress, cholinergic dysfunction, and inflammation in a mouse model of chronic cerebral hypoperfusion [53]. In a mouse model of brain I/R injury, the effects of nicorandil injection 15 min before the common carotid arteries occlusion were dose-dependent; a moderate number of pyknotic or shrunken nuclei were found in mice treated with lower doses of nicorandil (5 mg/kg), while pyknotic or shrunken nuclei were rarely found in mice treated with higher doses of nicorandil (10 mg/kg and 20 mg/kg) [63]. An injection of 10 mg/kg of nicorandil 30 min before MCAO resulted in a significant reduction in cerebral infarct volume [64]. In another study 5 mg/kg oral nicorandil administration once a day for three consecutive days after ischemia did not have a significant effect on neurobehavioral functions, count of neurons, non-neurons, and dead neurons, although it significantly reduced striatal infarct volumes [14]. In other investigations, normal and diabetic Wistar rats were given intraperitoneal nicorandil (5 mg/kg) for three days. They found a significant reduction in neurobehavioral impairments, cerebral infarct volume, and caspase enzyme levels compared to I/R control and I/R diabetic animals without affecting the animals' blood pressure (BP) [51]. Another study also reported that 5 mg/kg oral nicorandil administration once a day for three consecutive days after ischemia significantly decreased the neurological deficits and density of neuronal neighbors. It could not preserve the regular spatial distributions of M1 cortical neurons after MCAO. It also could not significantly improve motor function or reduce the ischemic lesion size [15]. Based on the mentioned preclinical studies, the effects of nicorandil on stroke outcomes can vary depending on the time of administration (see Table 1). Therefore, one major factor that needs attention is the temporal effects of nicorandil in stroke management.
Table 1.
Animal experiments with nicorandil in stroke.
| References | Animal model | Type of ischemia | Route of administration | Time of administration | Dose |
Evaluated parameters |
Main findings |
|---|---|---|---|---|---|---|---|
| [64] | male Wistar rats | MCAO (2 h) | Intraperitoneal injection | 30 min before MCAO | 10 mg/kg |
|
|
|
|
||||||
|
|
||||||
| [14] | Sprague–Dawley rats | MCAO (60 min) | oral gavage | for 3 consecutive days after MCAO | 5 mg/kg |
|
|
|
|
||||||
|
|
||||||
|
|
||||||
|
|
||||||
|
|
||||||
| Khastkhodaei et al. (unpublished) | male Sprague–Dawley rats | MCAO (60 min) | oral gavage | 30 min after MCAO | 1 (NIC1) and 3 (NIC3) mg/kg |
|
|
|
|
||||||
|
|
||||||
| [53] | male albino mice | Bilateral Common Carotid Artery Ligation or 2-Vessel Occlusion | oral gavage | once daily for 24 days | 2 and 4 mg/kg |
|
|
|
|
||||||
|
|
||||||
|
|
||||||
| [63] | Wild-type C57BL/6 mice |
Deep hypothermic low flow (DHLF) | Intraperitoneal injection | 15 min before occlusion of the CCAs | 5 mg/kg, 10 mg/kg, or 20 mg/kg) |
|
|
Western blot analysis
|
|
||||||
|
|
||||||
| [51] | male Wistar rats | MCAO (2 h) | Intraperitoneal injection | for 3 consecutive days after MCAO | 5 mg/kg |
|
|
|
|
||||||
|
|
||||||
|
|
||||||
| [15] | male Sprague–Dawley rats | MCAO (60 min) | oral gavage | for 3 consecutive days after MCAO | 5 mg/kg |
|
|
|
|
||||||
|
|
||||||
|
|
Abbreviations: AChE, Acetylcholinesterase; BLP, Brain lipid peroxidation; CAT, Catalase; CBF, Cerebral blood flow; CCA, Common carotid artery; CO, Cytochrome oxidase; CTRL, Control; GSH, Glutathione; MDA, Malondialdehyde; MPO, Myeloperoxidase; NIC, Nicorandil; SDH, Succinate dehydrogenase; SOD, Superoxide dismutase.
5. Therapeutic implications of nicorandil in ischemic stroke
The therapeutic implications and potential conjunct treatment of nicorandil in the context of ischemic injury are significant due to its dual mechanism of action as a NO donor and a K+ channel opener [21]. Given the similarity between ischemic injury in the heart and brain, nicorandil's potential to enhance cerebral blood flow (CBF) and reduce ischemic damage may have implications for stroke treatment [12]. Nicorandil may be beneficial as an adjunct therapy to complement standard stroke treatments. When administered alongside thrombolytic medications, which dissolve blood clots, nicorandil could help enhance the efficacy of clot-dissolving treatments [65]. Furthermore, nicorandil's potential to reduce inflammation, oxidative stress, and apoptosis could be considered alongside other neuroprotective strategies to minimize brain damage and improve outcomes [14]. In cases where reperfusion (restoration of blood flow) is a primary treatment goal, nicorandil's vasodilatory properties may aid in achieving optimal blood flow restoration in ischemic tissues and also reduce reperfusion injury through KATP channels [66].
6. Nicorandil effects on blood pressure
Hypertension is one of the modifiable risk factors for IS. Optimal blood pressure (BP) management differs in stroke patients. The 2018 guideline of the American Heart Association recommends patients eligible for tPA therapy should have their systolic BP below 185 mmHg and their diastolic BP below 110 mmHg [67]. However, hypotension is just as important as hypertension in stroke management. When cerebral ischemia occurs, studies have shown BP elevates even in patients who haven't had a history of hypertension [68]. This physiologic rise might be beneficial by improving CBF in the ischemic area [69]. A meta-regression analysis demonstrated that a mild reduction in BP may reduce mortality, however, significant falls or increases in BP are associated with worse clinical outcomes [70].
Nicorandil is a nitrate in chemical structure and causes vascular smooth muscle relaxation by increasing intracellular cyclic guanosine monophosphate (cGMP) which leads to NO release [28,71]. Nitrates like nicorandil cause peripheral venous dilatation leading to a reduction in preload and, therefore, coronary artery dilatation and lowering of systemic BP [28]. Nicorandil has a dual mechanism of action which can also act as a K+ channel opener that isn't related to its chemical structure [58]. Studies have demonstrated that non-nitrate K+ channel openers like BRL 34915 and pinacidil decrease systemic BP [72]. Unlike nitrates, nicorandil improves endothelial dysfunction [34]. Many studies have suggested that nicorandil has additional actions as an arterial KATP channel agonist, resulting in more “balanced” arterial and venous vasodilatation than nitrates [34] and the minimum effective doses of nicorandil produce its antianginal effects with a little hemodynamic side effect on BP, heart rate, or cardiac contractility [73].
A study that evaluated the effect of nicorandil on BP and CBF showed that nicorandil has a dual effect and is dose-dependent. The administration of 1 mg/kg nicorandil increased CBF (11.6 ± 3.6 vs. 0.5 ± 0.7%) in intact mice without reducing heart rate or systemic BP, while higher doses (5–10 mg/kg) resulted in a decrease in CBF (−6.6 ± 8.2 vs. 0.1 ± 1.1%), systemic BP (62 ± 12 vs. 76 ± 6 mmHg) and increased heart rate (523 ± 32 vs. 466 ± 18 beats/min) [74]. They concluded that the clinical dose of nicorandil can increase CBF without affecting systemic hemodynamics. Furthermore, intraperitoneal administration of nicorandil (5 mg/kg) for 3 days in a rat model of MCAO decreased neurobehavioral deficits, cerebral infarct volume, and caspase enzyme levels compared to Ischemic/Reperfusion control animals with no effect on BP (90 vs. 97 mmHg) [51].
Clinical trials on human subjects show nicorandil has a beneficial impact on mortality and morbidity in coronary artery disease patients [75] and post-AMI patients [76]. Previous studies showed that intracoronary injection of 4 mg/h for 24 h [77], intravenous of 12 mg [78], and 6 mg [79] nicorandil bolus exerted no significant effect on BP in ST-segment elevation myocardial infarction patients. While a bolus of 5 mg nicorandil, followed by 2 mg/h or 4 mg/h for 12 h continuously slightly decreased BP [80]. Supine BP was decreased equally and significantly after a single acute dose of 10, 20, and 30 mg nicorandil, with a peak after 4–6 h (10 mg: 21/-8 mmHg; 20 mg: 20/-9 mmHg; 30 mg: 29/-17 mmHg), and its hypotensive effect diminished after 24 h without a change in the heart rate [81]. In another study, nicorandil (2 or 4 mg/h) was given over 12 h after the percutaneous coronary intervention; systolic BP and systemic vascular resistance index were significantly reduced, suggesting that after a successful percutaneous coronary intervention, nicorandil reduced an afterload in patients with stable angina pectoris [82]. Therefore, these studies supported that a dose under 10 mg of intravenous nicorandil produced no deleterious effect on BP, but continuous intravenous administration of nicorandil led to a decrease in BP. The typical starting dose of nicorandil is 5–10 mg twice daily and can be up-titrated to 20 mg or a maximum of 30 mg twice daily [83]. Also, the cardioprotective dose of this drug utilized in animal models of I/R injury is considerably higher than the dose used in humans.
The use of nicorandil in the early post-stroke period may be effective in improving cerebral dysautoregulation in patients with hypertension. A study has demonstrated that nicorandil administration exerts beneficial effects on cerebral circulation in patients with chronic cerebral infarction by reducing mean arterial BP with increasing regional CBF [84]. It should be mentioned that the dose of nicorandil used should be able to increase CBF without affecting BP, intracranial pressures, or impairing cerebral autoregulation. A summary of these findings is presented in Table 2.
Table 2.
Clinical trials for the effects of nicorandil on blood pressure.
| References | Type of injury | Route of administration | Time of administration | Dose | Evaluated parameter | Main findings |
|---|---|---|---|---|---|---|
|
Patients with STEMI |
|
24 h | 4 mg/h |
|
|
|
|
|||||
| [85] | Patients with STEMI | Intravenous | 20–30 min before the PCI | 12 mg |
|
|
| [79] | Patients with STEMI | Intracoronary | After PCI | 6 mg |
|
|
| [80] | Patients with stable angina pectoris | Intravenous | 12 h after PCI | 2–4 mg/h |
|
|
| [86] | Hypertensive patients | Intravenous | 9 a.m. for 3 days | 10,20,30 mg |
|
|
| [87] | Patients with stable angina | Intravenous | After PCI | 5 mg bolus followed by 2 or 4 mg/h for 12 h |
|
|
Abb Abbreviations: AMI, Acute myocardial infarction; BP, Blood pressure; CTRL, Control group; IHD, Ischemic heart disease; NIC, Nicorandil treated group; PCI, Percutaneous coronary intervention; PPCI, Primary percutaneous coronary intervention; STEMI; ST-elevation myocardial infarction.
7. Conclusion
In this review, we have provided evidence that KATP channels not only have neuroprotective effects through preserving the brain's energy but also through modulating the immune response following IS. Nicorandil is a K+ channel opener and NO donor. Opening of mitochondrial KATP channels by nicorandil can protect the brain penumbra during ischemia by reducing intracellular Ca2+ levels and preventing apoptosis and cell death. The K+ channels induce hyperpolarization and can decrease neuronal activity and therefore, preserve the brain's energy.
Nicorandil has demonstrated encouraging outcomes in reducing inflammation, cerebral edema, and preventing apoptosis, factors that can have a substantial impact on the outcomes of stroke. These findings which support that nicorandil may improve overall stroke outcomes are mainly from animal studies, therefore further trials are needed to better evaluate its efficacy in IS.
Nicorandil's effects on BP need further attention in stroke management. Based on previous studies, it seems that the right dose of nicorandil can improve CBF without affecting systemic hemodynamics. This is due to the NO pathway and K+ channel opening mechanism. Significant falls in blood pressure can lead to worse stroke outcomes as it can disrupt blood perfusion. Therefore, it's crucial to consider nicorandil's role in this matter and choose the appropriate dosage for stroke management. However, the use of nicorandil in managing stroke is not yet a standard practice and further trials are needed to establish its safety and effectiveness in this context.
These findings bring new insights into how nicorandil can potentially have neuroprotective effects and promising outcomes that may change future prognosis and IS management.
Funding
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Data availability
No data was used for the research described in the article.
CRediT authorship contribution statement
Maryam Owjfard: Writing – review & editing, Writing – original draft, Software, Methodology, Investigation, Formal analysis, Data curation, Conceptualization. Negin Rahmani: Writing – review & editing, Writing – original draft. Arashk Mallahzadeh: Writing – review & editing, Writing – original draft. Mahnaz Bayat: Writing – review & editing, Writing – original draft, Formal analysis. Afshin Borhani-Haghighi: Writing – review & editing, Writing – original draft, Data curation. Farzaneh Karimi: Writing – review & editing, Writing – original draft, Methodology. Mohammad Reza Namavar: Writing – review & editing, Visualization, Validation, Supervision, Project administration, Methodology, Data curation, Conceptualization.
Declaration of competing interest
The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: The authors declare that they have no competing interests. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- 1.Owjfard M., et al. Effect of dimethyl fumarate on the motor function and spatial arrangement of primary motor cortical neurons in the sub-acute phase of stroke in a rat model. 2021;30(4) doi: 10.1016/j.jstrokecerebrovasdis.2021.105630. [DOI] [PubMed] [Google Scholar]
- 2.Owjfard M., et al. vol. 58. 2022. (Therapeutic Effects of Dimethyl Fumarate on the Rat Model of Brain Ischemia). [Google Scholar]
- 3.Siddiqi A.Z., Wadhwa A. Treatment of acute stroke: current practices and future horizons. Cardiovasc. Revascularization Med. 2022;49:56–65. doi: 10.1016/j.carrev.2022.11.012. [DOI] [PubMed] [Google Scholar]
- 4.Broderick J.P., et al. Endovascular therapy after intravenous t-PA versus t-PA alone for stroke. N. Engl. J. Med. 2013;368(10):893–903. doi: 10.1056/NEJMoa1214300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Grotta J.C., Hacke W. Stroke neurologist's perspective on the new endovascular trials. Stroke. 2015;46(6):1447–1452. doi: 10.1161/STROKEAHA.115.008384. [DOI] [PubMed] [Google Scholar]
- 6.Polk S.R., et al. Contraindications with recombinant tissue plasminogen activator (rt-PA) in acute ischemic stroke population. Neurol. Psychiatr. Brain Res. 2018;27:6–11. [Google Scholar]
- 7.Saqqur M., et al. Site of arterial occlusion identified by transcranial Doppler predicts the response to intravenous thrombolysis for stroke. Stroke. 2007;38(3):948–954. doi: 10.1161/01.STR.0000257304.21967.ba. [DOI] [PubMed] [Google Scholar]
- 8.Campbell B.C., et al. Endovascular stent thrombectomy: the new standard of care for large vessel ischaemic stroke. Lancet Neurol. 2015;14(8):846–854. doi: 10.1016/S1474-4422(15)00140-4. [DOI] [PubMed] [Google Scholar]
- 9.Chia N.H., et al. Determining the number of ischemic strokes potentially eligible for endovascular thrombectomy: a population-based study. Stroke. 2016;47(5):1377–1380. doi: 10.1161/STROKEAHA.116.013165. [DOI] [PubMed] [Google Scholar]
- 10.Furlan A.J. Endovascular therapy for stroke–it’s about time. N. Engl. J. Med. 2015;372(24):2347–2349. doi: 10.1056/NEJMe1503217. [DOI] [PubMed] [Google Scholar]
- 11.Josephson S.A., Kamel H. The acute stroke care revolution: enhancing access to therapeutic advances. JAMA. 2018;320(12):1239–1240. doi: 10.1001/jama.2018.11122. [DOI] [PubMed] [Google Scholar]
- 12.Widimský P., et al. Acute myocardial infarction and acute stroke: what are the differences? Focus on reperfusion therapy. Cor Vasa. 2013;55(2):e111–e116. [Google Scholar]
- 13.Treese N., Erbel R., Meyer J. Acute hemodynamic effects of nicorandil in coronary artery disease. J. Cardiovasc. Pharmacol. 1992;20(Suppl 3):S52–S56. doi: 10.1097/00005344-199206203-00010. [DOI] [PubMed] [Google Scholar]
- 14.Owjfard M., et al. Effects of nicorandil on neurobehavioral function, BBB integrity, edema and stereological parameters of the brain in the sub-acute phase of stroke in a rat model. J. Biosci. 2020;45 [PubMed] [Google Scholar]
- 15.Owjfard M., et al. Effect of nicorandil on the spatial arrangement of primary motor cortical neurons in the sub-acute phase of stroke in a rat model. J. Chem. Neuroanat. 2021;117 doi: 10.1016/j.jchemneu.2021.102000. [DOI] [PubMed] [Google Scholar]
- 16.Zhu F., et al. Protective effects of nicorandil against cerebral injury in a swine cardiac arrest model. Exp. Ther. Med. 2018;16(1):37–44. doi: 10.3892/etm.2018.6136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Geshi E., et al. The role of ATP-sensitive potassium channels in the mechanism of ischemic preconditioning. J. Cardiovasc. Pharmacol. 1999;34(3):446–453. doi: 10.1097/00005344-199909000-00020. [DOI] [PubMed] [Google Scholar]
- 18.Matsubara T., et al. Three minute, but not one minute, ischemia and nicorandil have a preconditioning effect in patients with coronary artery disease. J. Am. Coll. Cardiol. 2000;35(2):345–351. doi: 10.1016/s0735-1097(99)00539-2. [DOI] [PubMed] [Google Scholar]
- 19.Mizumura T., Gross G. Infarct-reducing effect of nicorandil, an ATP-sensitive potassium channel opener-nitrate, is reversed by glibenclamide, but not blocked by methylene glue in dogs: comparison with nitroglycerin. Circulation. 1995;92(I):I–253. [Google Scholar]
- 20.Markham A., Plosker G.L., Goa K.L. Nicorandil. Drugs. 2000;60(4):955–974. doi: 10.2165/00003495-200060040-00007. [DOI] [PubMed] [Google Scholar]
- 21.Ahmed L.A. Nicorandil: a drug with ongoing benefits and different mechanisms in various diseased conditions. Indian J. Pharmacol. 2019;51(5):296. doi: 10.4103/ijp.IJP_298_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Haddad G., Jiang C. O2 deprivation in the central nervous system: on mechanisms of neuronal response, differential sensitivity and injury. Prog. Neurobiol. 1993;40(3):277–318. doi: 10.1016/0301-0082(93)90014-j. [DOI] [PubMed] [Google Scholar]
- 23.Lipton P. Ischemic cell death in brain neurons. Physiol. Rev. 1999;79(4):1431–1568. doi: 10.1152/physrev.1999.79.4.1431. [DOI] [PubMed] [Google Scholar]
- 24.Bickler P.E., Buck L.T. Adaptations of vertebrate neurons to hypoxia and anoxia: maintaining critical Ca2+ concentrations. J. Exp. Biol. 1998;201(8):1141–1152. doi: 10.1242/jeb.201.8.1141. [DOI] [PubMed] [Google Scholar]
- 25.Kristián T., Siesjö B.K. Calcium-related damage in ischemia. Life Sci. 1996;59(5–6):357–367. doi: 10.1016/0024-3205(96)00314-1. [DOI] [PubMed] [Google Scholar]
- 26.Nicholls D.G., Budd S.L. Mitochondria and neuronal survival. Physiol. Rev. 2000;80(1):315–360. doi: 10.1152/physrev.2000.80.1.315. [DOI] [PubMed] [Google Scholar]
- 27.Hansen A.J. Effect of anoxia on ion distribution in the brain. Physiol. Rev. 1985;65(1):101–148. doi: 10.1152/physrev.1985.65.1.101. [DOI] [PubMed] [Google Scholar]
- 28.Ahmed L.A. Nicorandil: a drug with ongoing benefits and different mechanisms in various diseased conditions. Indian J. Pharmacol. 2019;51(5):296–301. doi: 10.4103/ijp.IJP_298_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ballanyi K. Protective role of neuronal KATP channels in brain hypoxia. J. Exp. Biol. 2004;207(18):3201–3212. doi: 10.1242/jeb.01106. [DOI] [PubMed] [Google Scholar]
- 30.Luhmann H.J., Heinemann U. Hypoxia-induced functional alterations in adult rat neocortex. J. Neurophysiol. 1992;67(4):798–811. doi: 10.1152/jn.1992.67.4.798. [DOI] [PubMed] [Google Scholar]
- 31.Mourre C., et al. Antidiabetic sulfonylureas: localization of binding sites in the brain and effects on the hyperpolarization induced by anoxia in hippocampal slices. Brain Res. 1989;486(1):159–164. doi: 10.1016/0006-8993(89)91288-2. [DOI] [PubMed] [Google Scholar]
- 32.Sun H.-s., Feng Z.-p. Neuroprotective role of ATP-sensitive potassium channels in cerebral ischemia. Acta Pharmacol. Sin. 2013;34(1):24–32. doi: 10.1038/aps.2012.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Feng C., et al. Effect of nicorandil administration on myocardial microcirculation during primary percutaneous coronary intervention in patients with acute myocardial infarction. Postępy w Kardiologii Interwencyjnej= Advances in Interventional Cardiology. 2018;14(1):26. doi: 10.5114/aic.2018.74352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tarkin J.M., Kaski J.C. Vasodilator therapy: nitrates and nicorandil. Cardiovasc. Drugs Ther. 2016;30(4):367–378. doi: 10.1007/s10557-016-6668-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhao X.-T., Zhang C.-F., Liu Q.-J. Meta-analysis of Nicorandil effectiveness on myocardial protection after percutaneous coronary intervention. BMC Cardiovasc. Disord. 2019;19(1):144. doi: 10.1186/s12872-019-1071-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bajgar R., et al. Identification and properties of a novel intracellular (mitochondrial) ATP-sensitive potassium channel in brain. J. Biol. Chem. 2001;276(36):33369–33374. doi: 10.1074/jbc.M103320200. [DOI] [PubMed] [Google Scholar]
- 37.Héron-Milhavet L., et al. Protection against hypoxic–ischemic injury in transgenic mice overexpressing Kir6. 2 channel pore in forebrain. Mol. Cell. Neurosci. 2004;25(4):585–593. doi: 10.1016/j.mcn.2003.10.012. [DOI] [PubMed] [Google Scholar]
- 38.Kis B., et al. Diazoxide induces delayed pre‐conditioning in cultured rat cortical neurons. J. Neurochem. 2003;87(4):969–980. doi: 10.1046/j.1471-4159.2003.02072.x. [DOI] [PubMed] [Google Scholar]
- 39.Hosseini S.M., et al. Preconditioned neurons with NaB and nicorandil, a favorable source for stroke cell therapy. J. Cell. Biochem. 2018;119(12):10301–10313. doi: 10.1002/jcb.27372. [DOI] [PubMed] [Google Scholar]
- 40.Bi Y.Y., Quan Y. PirB inhibits axonal outgrowth via the PI3K/Akt/mTOR signaling pathway. Mol. Med. Rep. 2018;17(1):1093–1098. doi: 10.3892/mmr.2017.7930. [DOI] [PubMed] [Google Scholar]
- 41.Othman F.A., Tan S.C. Preconditioning strategies to enhance neural stem cell-based therapy for ischemic stroke. Brain Sci. 2020;10(11):893. doi: 10.3390/brainsci10110893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Orellana-Urzúa S., et al. Pathophysiology of ischemic stroke: role of oxidative stress. Curr. Pharmaceut. Des. 2020;26(34):4246–4260. doi: 10.2174/1381612826666200708133912. [DOI] [PubMed] [Google Scholar]
- 43.Wu L., et al. The neuroprotection conferred by activating the mitochondrial ATP-sensitive K+ channel is mediated by inhibiting the mitochondrial permeability transition pore. Neurosci. Lett. 2006;402(1–2):184–189. doi: 10.1016/j.neulet.2006.04.001. [DOI] [PubMed] [Google Scholar]
- 44.Bai Y.P., Han L.S. Nicorandil alleviated cardiac hypoxia/reoxygenation-induced cytotoxicity via upregulating ketone body metabolism and ACAT1 activity. KOREAN J. PHYSIOL. PHARMACOL. 2019;23(1):37–45. doi: 10.4196/kjpp.2019.23.1.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yamada S., et al. Nicorandil reduces intracellular calcium accumulation Evoked by oxygen glucose deprivation in rat hippocampal CA1 pyramidal cells. Anesthesiology. 2005;103(A188):A188. [Google Scholar]
- 46.Dostovic Z., et al. Brain edema after ischaemic stroke. Med. Arch. 2016;70(5):339. doi: 10.5455/medarh.2016.70.339-341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Badaut J., Plesnila N. Academic Press; 2017. Brain Edema: from Molecular Mechanisms to Clinical Practice. [Google Scholar]
- 48.Pirici I., et al. Inhibition of aquaporin-4 improves the outcome of ischaemic stroke and modulates brain paravascular drainage pathways. Int. J. Mol. Sci. 2018;19(1):46. doi: 10.3390/ijms19010046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhu F., et al. Protective effects of nicorandil against cerebral injury in a swine cardiac arrest model. Exp. Ther. Med. 2018;16(1):37–44. doi: 10.3892/etm.2018.6136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Dong Y.-F., et al. Potential role of microRNA-7 in the anti-neuroinflammation effects of nicorandil in astrocytes induced by oxygen-glucose deprivation. J. Neuroinflammation. 2016;13(1):60. doi: 10.1186/s12974-016-0527-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Pithadia A.B., Panchal S.S., Patel D.J. Neuroprotective effects of potassium channel openers on cerebral ischemia–reperfusion injury in diabetic rats. Bull. Fac. Pharm. Cairo Univ. 2017;55(1):95–100. [Google Scholar]
- 52.Tajima M., et al. Nicorandil enhances the effect of endothelial nitric oxide under hypoxia-reoxygenation: role of the KATP channel. Eur. J. Pharmacol. 2008;579(1–3):86–92. doi: 10.1016/j.ejphar.2007.10.074. [DOI] [PubMed] [Google Scholar]
- 53.Gupta S., Singh P., Sharma B. Neuroprotective effects of nicorandil in chronic cerebral hypoperfusion-induced vascular dementia. J. Stroke Cerebrovasc. Dis. 2016;25(11):2717–2728. doi: 10.1016/j.jstrokecerebrovasdis.2016.07.023. [DOI] [PubMed] [Google Scholar]
- 54.Benakis C., et al. The role of microglia and myeloid immune cells in acute cerebral ischemia. Front. Cell. Neurosci. 2015;8:461. doi: 10.3389/fncel.2014.00461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zheng Z., Yenari M.A. Post-ischemic inflammation: molecular mechanisms and therapeutic implications. Neurol. Res. 2004;26(8):884–892. doi: 10.1179/016164104X2357. [DOI] [PubMed] [Google Scholar]
- 56.Yang G.-Y., et al. Tumor necrosis factor alpha expression produces increased blood–brain barrier permeability following temporary focal cerebral ischemia in mice. Mol. Brain Res. 1999;69(1):135–143. doi: 10.1016/s0169-328x(99)00007-8. [DOI] [PubMed] [Google Scholar]
- 57.Chaturvedi M., Kaczmarek L. Mmp-9 inhibition: a therapeutic strategy in ischemic stroke. Mol. Neurobiol. 2014;49(1):563–573. doi: 10.1007/s12035-013-8538-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ahmed A.A., et al. Dimethyl fumarate interferes with MyD88-dependent toll-like receptor signalling pathway in isoproterenol-induced cardiac hypertrophy model. J. Pharm. Pharmacol. 2018;70(11):1521–1530. doi: 10.1111/jphp.13000. [DOI] [PubMed] [Google Scholar]
- 59.Zhao A.P., et al. Nicorandil inhibits inflammasome activation and Toll-like receptor-4 signal transduction to protect against oxygen-glucose deprivation-induced inflammation in BV-2 cells. CNS Neurosci. Ther. 2014;20(2):147–153. doi: 10.1111/cns.12178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Peng R., Yang X., Liang X. Nicorandil effects on platelet function, Hs-CRP, MMP-9 and myocardial antioxidation in patients with unstable angina. Exp. Ther. Med. 2019;18(4):3095–3099. doi: 10.3892/etm.2019.7918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Teshima Y., et al. Nicorandil prevents oxidative stress-induced apoptosis in neurons by activating mitochondrial ATP-sensitive potassium channels. Brain Res. 2003;990:45–50. doi: 10.1016/s0006-8993(03)03383-3. [DOI] [PubMed] [Google Scholar]
- 62.Frampton J., Buckley M.M., Fitton A., Nicorandil A review of its pharmacology and therapeutic efficacy in angina pectoris. Drugs. 1992;44(4):625–655. doi: 10.2165/00003495-199244040-00008. [DOI] [PubMed] [Google Scholar]
- 63.Yu D., et al. Neuroprotective effect of nicorandil through inhibition of apoptosis by the PI3K/Akt1 pathway in a mouse model of deep hypothermic low flow. J. Neurol. Sci. 2015;357(1–2):119–125. doi: 10.1016/j.jns.2015.07.010. [DOI] [PubMed] [Google Scholar]
- 64.Wang P., et al. Effect of nicorandil on infarct volume and marker enzyme activity in mitochondria of rats with cerebral ischemia/reperfusion injury. Neural Regeneration Research. 2008;3(5):501–504. [Google Scholar]
- 65.Yu S.-H., Li L., Zhang S. Clinical effect of intravenous thrombolysis combined with nicorandil therapy in patients with acute ST-segment elevation myocardial infarction. Trop. J. Pharmaceut. Res. 2016;15(6):1335–1339. [Google Scholar]
- 66.Lee H.C., et al. Effect of intra-coronary nicorandil administration prior to reperfusion in acute ST segment elevation myocardial infarction. Circ. J. 2008;72(9):1425–1429. doi: 10.1253/circj.cj-08-0212. [DOI] [PubMed] [Google Scholar]
- 67.Powers W.J., et al. Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2018;49(3):e46–e99. doi: 10.1161/STR.0000000000000158. [DOI] [PubMed] [Google Scholar]
- 68.Wallace J., Levy L. Blood pressure after stroke. JAMA. 1981;246(19):2177–2180. [PubMed] [Google Scholar]
- 69.Weiss A., et al. Change in systolic blood pressure during stroke, functional status, and long-term mortality in an elderly population. Am. J. Hypertens. 2016;29(4):432–438. doi: 10.1093/ajh/hpv118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Geeganage C.M., Bath P.M. Relationship between therapeutic changes in blood pressure and outcomes in acute stroke: a metaregression. Hypertension. 2009;54(4):775–781. doi: 10.1161/HYPERTENSIONAHA.109.133538. [DOI] [PubMed] [Google Scholar]
- 71.Endoh M., Taira N. Relationship between relaxation and cyclic GMP formation caused by nicorandil in canine mesenteric artery. N. Schmied. Arch. Pharmacol. 1983;322(4):319–321. doi: 10.1007/BF00508349. [DOI] [PubMed] [Google Scholar]
- 72.Taira N. Similarity and dissimilarity in the mode and mechanism of action between nicorandil and classical nitrates: an overview. J. Cardiovasc. Pharmacol. 1987;10:S1–S9. [PubMed] [Google Scholar]
- 73.Kinoshita M., Sakai K. Pharmacology and therapeutic effects of nicorandil. Cardiovasc. Drugs Ther. 1990;4(4):1075–1088. doi: 10.1007/BF01856503. [DOI] [PubMed] [Google Scholar]
- 74.Kotoda M., et al. Nicorandil increased the cerebral blood flow via nitric oxide pathway and ATP-sensitive potassium channel opening in mice. J. Anesth. 2018;32(2):244–249. doi: 10.1007/s00540-018-2471-2. [DOI] [PubMed] [Google Scholar]
- 75.Horinaka S., et al. Effects of nicorandil on cardiovascular events in patients with coronary artery disease in the Japanese Coronary Artery Disease (JCAD) study. Circ. J. 2010;3:503–509. doi: 10.1253/circj.cj-09-0649. [DOI] [PubMed] [Google Scholar]
- 76.Sakata Y., et al. Oral treatment with nicorandil at discharge is associated with reduced mortality after acute myocardial infarction. J. Cardiol. 2012;59(1):14–21. doi: 10.1016/j.jjcc.2011.08.001. [DOI] [PubMed] [Google Scholar]
- 77.Pi S.-F., et al. Effect of sequential nicorandil on myocardial microcirculation and short-term prognosis in acute myocardial infarction patients undergoing coronary intervention. J. Thorac. Dis. 2019;11(3):744–752. doi: 10.21037/jtd.2019.02.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ishii H., et al. Impact of a single intravenous administration of nicorandil before reperfusion in patients with ST-segment–elevation myocardial infarction. Circulation 2005;. 112(9):1284. doi: 10.1161/CIRCULATIONAHA.104.530329. [DOI] [PubMed] [Google Scholar]
- 79.Wang Z., et al. The effect on myocardial perfusion and clinical outcome of intracoronary nicorandil injection prior to percutaneous coronary intervention in ST-segment elevation myocardial infarction. Zhonghua Xinxueguanbing Zazhi. 2017;45(1):26. doi: 10.3760/cma.j.issn.0253-3758.2017.01.006. [DOI] [PubMed] [Google Scholar]
- 80.Sato S., et al. Effect of nicorandil administration on cardiac burden and cardio-ankle vascular index after coronary intervention. Heart Ves. 2020;35(12):1664–1671. doi: 10.1007/s00380-020-01650-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Leonetti G., et al. Nicorandil, a new vasodilator drug, in patients with essential hypertension. J. Hypertens. Suppl. 1989;7(6):S292–S293. doi: 10.1097/00004872-198900076-00142. [DOI] [PubMed] [Google Scholar]
- 82.Sato S., et al. PS 02-09 Effect of nicorandil administration on arterial stiffness monitored with the cardio-ankle vascular index (CAVI) after coronary intervention. J. Hypertens. 2016;34:e106. [Google Scholar]
- 83.Tarkin J.M., Kaski J.C. Nicorandil and long-acting nitrates: vasodilator therapies for the management of chronic stable angina pectoris. European cardiology. 2018;13(1):23–28. doi: 10.15420/ecr.2018.9.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kobayashi S., et al. Effects of nicorandil on regional cerebral blood flow in patients with chronic cerebral infarction. Preliminary communication. Arzneimittelforschung. 1992;42(9):1086–1089. [PubMed] [Google Scholar]
- 85.Ishii H., et al. Impact of a single intravenous administration of nicorandil before reperfusion in patients with ST-segment–elevation myocardial infarction. Circulation. 2005;112(9):1284–1288. doi: 10.1161/CIRCULATIONAHA.104.530329. [DOI] [PubMed] [Google Scholar]
- 86.Leonetti G., et al. Nicorandil, a new vasodilator drug, in patients with essential hypertension. J. Hypertens. Suppl.: Official Journal of the International Society of Hypertension. 1989;7(6):S292–S293. doi: 10.1097/00004872-198900076-00142. [DOI] [PubMed] [Google Scholar]
- 87.Sato S., et al. Effect of nicorandil administration on cardiac burden and cardio-ankle vascular index after coronary intervention. Heart Ves. 2020;35:1664–1671. doi: 10.1007/s00380-020-01650-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No data was used for the research described in the article.

Evaluated parameters