IMR Press / RCM / Volume 22 / Issue 3 / DOI: 10.31083/j.rcm2203073
Open Access Review
Management of myocardial infarction with Nonobstructive Coronary Arteries (MINOCA): a subset of acute coronary syndrome patients
Show Less
1 Department of Cardiology, Toulouse University Hospital, 31059 Toulouse, France
2 Faculty of Medicine, Holy Spirit University of Kaslik, 446 Jounieh, Lebanon
3 Faculty of Pharmacy, Lebanese University, 13-5033 Beirut, Lebanon
*Correspondence: roncalli.j@chu-toulouse.fr (Jerome Roncalli)
Academic Editor: Leonardo De Luca
Rev. Cardiovasc. Med. 2021, 22(3), 625–634; https://doi.org/10.31083/j.rcm2203073
Submitted: 30 April 2021 | Revised: 4 June 2021 | Accepted: 11 June 2021 | Published: 24 September 2021
Copyright: © 2021 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license (https://creativecommons.org/licenses/by/4.0/).
Abstract

Myocardial infarction with non-obstructive coronary artery disease (MINOCA) represents a significant proportion (up to 15%) of acute myocardial infarction (AMI) population. MINOCA is diagnosed in patients who fullfilled the fourth universal definition of AMI in the absence of significant obstructive coronary artery disease on coronary angiography. MINOCA is a group of heterogeneous diseases with different pathophysiological mechanisms requiring multimodality imaging. Left ventriculography, cardiac magnetic resonance imaging and intra-coronary imaging (IVUS, OCT) are useful tools playing a pivotal role in the diagnostic work-up. There are no standard guidelines on the management of MINOCA patients and the therapeutic approach is personalized, thereby detecting the underlying aetiology is fundamental to initiate an early appropriate cause-targeted therapy.

Keywords
MINOCA
Management
Myocardial infarction
Atherosclerosis
1. Introduction

Myocardial infarction with non-obstructive coronary artery disease (MINOCA) represents a significant proportion (up to 15%) of acute myocardial infarction (AMI) population [1, 2]. This term on top of others such as INOCA (ischemia and no obstructive coronary artery disease) [3], MINCA (myocardial infarction with normal coronary arteries) [4], TP-NOCA (troponin-positive non-obstructive coronary arteries) and ACSNNOCA (acute coronary syndrome with normal or near normal coronary arteries) [5] were used to describe a sole clinical entity incorporating myocardial damage in the absence of significant obstructive coronary artery disorder. While MINOCA includes a group of miscellaneous diseases, the underlying aetiology and pathophysiological mechanisms remain unclear in large patient series [6]. Coronary angiography is the crucial exam for making the diagnosis of MINOCA which requires to exclude the presence of significant coronary artery stenosis (50% luminal narrowing) and an alternative cause of the acute presentation in the setting of AMI [6, 7]. The fourth universal definition of myocardial infarction requests a rise and/or fall of high-sensitive cardiac troponine (hs-CT) with one value above the 99-percentile upper reference limit [7]. Compared to CAD-AMI (significant coronary artery disease- acute myocardial infarction) population, MINOCA patients were younger and have lower prevalence of cardiovascular risk factors including dyslipidemia, hypertension, diabetes mellitus, and smoking [8, 9]. Even though, MINOCA patients were preponderantly females and previous studies showed that the incidence of MINOCA in women presenting with AMI is twofolds higher than that of men [8, 9]. Atherosclerosis has a limited burden in MINOCA patients unlike CAD-AMI population, thereby playing a minor role in the pathophysiology of AMI in MINOCA cases. Noteworthy that according to aforementionned MINOCA diagnostic criterias, clinical entities resulting in increased level of hs-CT such as stroke, pulmonary embolism, sepsis, adult respiratory distress syndrome and end-stage renal failure must be excluded in the work-up [6, 7]. MINOCA clinically manifests as no ST-elevation myocardial infarction rather than ST-elevation myocardial infarction (2/3 vs 1/3 of cases) [10]. The long-term prognosis is usually better but not trivial among MINOCA patients compared to those with CAD-AMI [10, 11]. Reduced left ventricle ejection fraction [11, 12, 13], increased C-reactive protein [14], elevated level of hs-CT [15, 16], female sex [12, 17] and mild three-vessels or left main atherosclerosis (30–50% luminal stenosis) [14] are independant predictors of adverse events in MINOCA patients. Herein, we focus on the therapeutic approach and management of MINOCA: a specific subset of acute coronary syndrome population.

2. Causes of MINOCA

The main causes of MINOCA could be divided into two groups: ischemic and non-ischemic.

2.1 The ischemic causes

The ischemic causes include spontaneous coronary artery dissection (SCAD), plaque disruption, coronary spasm, microvascular dysfunction, coronary thrombus or embolism and supply-demand mismatch.

Coronary artery dissection is a tear separating the inner layers of the coronary artery from the outer layers not always revealed by coronary angiography [3, 18]. However, intramural haematoma without intimal tear is detected by intracoronary imaging in some cases [19]. Fibromuscular dysplasia is commonly found in other vessels of SCAD-MINOCA patients [20]. It is more often in women accounting for 25% of all acute coronary syndromes in women aged below 50-year-old [21]. The impacts of hormonal changes, pregnancy and delivery on vascular wall (intima-media) were implicated [21]. In SCAD, the external compression of coronary true lumen results in myocardial infarction or MINOCA. It is the consequence of blood accumulation in the tunica media after an endothelium-intimal tear formation “inside-out mechanism” or microvessel rupture in the vascular wall “outside-in mechanism” [22, 23]. A recent OCT based study found that the absence of fenestration in SCAD increases the pressure in false lumen and subsequently the external compression of the true lumen [24].

MINOCA accounts for up to 20% of type 1 AMI enrolling plaque rupture and erosion [6, 25] which were detected by IVUS among 40% of MINOCA cases [26, 27]. Thromboembolism, superadded coronary spasm, thrombosis and an association of these processes are the main mechanisms of myocardial necrosis in MINOCA patients with plaque rupture. Plaques with thin fibroatheromatous cap (<65 μm) and infiltrated by macrophages are at risk for rupturing [28]. Activation of the sympathetic nervous system is the main trigger for plaque rupture while hypercoagulable state amplifies the thrombotic response [29]. Plaque erosion results in thrombus formation without rupture, thereby the internal elastic lamina and underlying media are intacts unike plaque rupture [30]. Thus, spontaneous coronary thrombolysis has been proposed as an internal protective mechanism avoiding thrombus formation in case of plaque rupture or erosion [26].

The prevalence of coronary artery spasm defined as a reversible diffuse or focal vascular smooth muscle hypersensitivity to vasocontrictive endogenous or exogenous stimuli varies between 3 to 95% of MINOCA cases [31, 32]. It is well known that modifications in coronary vasomotor tone participate in the pathogenesis of myocardial infarction. Then, it has been demonstrated that a mutation (V734I) in the nucleotide binding domain binding 1 of ABCC9 affects the susceptibility to AMI by altering the vasomotor response to intracellular nucelotides [33]. Coronary microvascular spasm or dysfunction altering myocardial perfusion and contractility has been found in 25% of MINOCA patients [34]. It induced transient myocardial ischemia with electrical modifications (ST segment), clinical manifestation (angina) and troponin elevations without angiographic reduction in coronary lumen [35, 36, 37]. Despite the lack of epicardial obstructive disease, there can be microvascular obstruction which is a cause of MINOCA.

Acquired thrombotic disorders (antiphospholipid syndrome, myeloproliferative diseases), hereditary thrombotic disorders (Protein C, S and facteur V Leiden deficiencies) and predisposing hypercoagulable states (atrial fibrillation, cardiac tumor, valvular vegetations or calcifications) result in coronary thromboembolism [10]. Structural heart disease like right-left shunts, patent forman ovale, atrial septal defect and coronary fistula may lead to paradoxical embolism which is relatively a rare cause of MINOCA [38, 39, 40]. Lastly, conditions provoking oxygen supply-demand mismatch by either increasing oxygen demand through promoting systolic wall tension, myocardial contractility and heart rate or reducing oxygen supply through decreasing coronary blood flow and oxygen-carrying capacity are responsible for type 2 AMI [41, 42, 43].

2.2 The non-ischemic causes

The non-ischemic causes include myocarditis, tako-tsubo cardiomyopathy, hypertensive heart disease, tachyarrhythmias, cardiotoxins or chemotherapeutic agents and cardiomyopathies. Myocarditis are likely to present as acute coronary syndrome accounting for 33% of MINOCA cases [44] especially when parvovirus B19 is the underlying causative agent [45]. Parvovirus B19 has predilection to endothelial cells due to blood group P antigen inducing intense vasocontriction of coronary microciculation and subsequent ST segment elevation in the setting of myocardial inflammation [46, 47]. This finding was confirmed by Yilmaz et al. [47] who showed an epicardial spasm limited to the distal part of coronary artery after the injection of acetylcholine in MINOCA patients with myocarditis [46, 47]. Viral infections with adenoviruses, coxsackie virus, parvovirus B19 and human herpes virus 6 are the most common cause of myocarditis [45].

Toxins, drugs, chemotherapeutic agents, auto-immune (sarcoidosis, lupus) and endocrine diseases are another causes of myocarditis [48, 49, 50]. Myocardial involvement during autoimmune disorders could be the sole clinical presentation or a part of systemic reaction [51]. Early diagnosis of myocarditis in the context of MINOCA is important for its prognostic value and to establish an appropriate treatment [52]. Indeed, a poor prognosis was attributed to eosinophilic myocarditis, cardiac sarcoidosis and giant cell myocarditis [52].

Tako-tsubo cardiomyopathy known as stress-induced reversible cardiomyopathy generally affects post-menopausal women. The exact pathophysiological mechanisms remain unclear, thereby several hypotheses such as diffuse spasm, spontaneous coronary thrombus and lysis, cathecholamine-induced myocardial stunning have been proposed. However, coronary microvascular dysfunction is a common finding among tako-tsubo syndromes [53].

Lastly, illicit drug use such as cocaine and MDMA can also result in MINOCA.

2.3 Novelties in MINOCA causes

In the era of COVID-19 (coronavirus disease 2019) pandemic, COVID-19 infection was found as a potential causative agent for MINOCA or ACSNNOCA. COVID-19 positive patients are likely to develop tako-tsubo cardiomyopathy (stressful situation), myocarditis (viral inflammation), AMI (thrombo-embolism, vasospasm and plaque rupture) and non-cardiac conditions such as pulmonary embolism and sepsis [54]. A pro-thrombotic effect ensuing from local and systemic inflammatory reactions, endothelial cells infection and cytokines storm has been currently attributed to COVID-19 [55, 56, 57].

Recently, a correlation between MINOCA and myocardial bridge has been hypothesized by our group via a large observational cohort [58]. In fact, myocardial bridge has the potential to activate and enhance the all suggested pathophysiological mechanisms of MINOCA. The mechanical compression called systolic milking effect of MB predisposed to atherosclerotic plaque rupture [59] or contributed to SCAD [60]. Moreover, the tunneled coronary segments are prompt to vasospasm with hyper-contractility response to vasoconstrictive agents [61]. The continuous transient dynamic compression of vascular wall results in increasing the shear wall stress which harm endothelial cells [62, 63]. Data from literature showed that myocardial infarction could be a pertinent clinical manifestation of myocardial bridge with milking effect [64, 65, 66] and recent published study revealed a significantly higher angiographic prevalence of myocardial bridge in MINOCA rather than CAD-AMI (coronary artery disease acute myocardial infarction) populations [58, 66]. It is worthy to mention that myocardial bridge is associated with proximal atherosclerotic plaque despite sparing of the bridge, so it remains to be seen if the bridge and/or combination of bridge and non-obstructive CAD result in MINOCA.

3. Management of patients suspected of MINOCA

MINOCA should be considered as «working diagnosis» rather than «true diagnosis», thereby performing further diagnostic evaluation is fundamental. Coronary angiography is urgently recommended in patients with ST-segment elevation myocardial infarction while it could be delayed according to cardiovascular risk stratification in those with No ST-segment elevation myocardial infarction. Screening for AMI mechanical complications, transthoracic echocardiography is routinely performed before undergoing cardiac catheterization laboratory. In the absence of significant coronary artery stenosis on coronary angiogram, C-MRI should be performed to differentiate ischemic from non-ischemic causes. According to clinical context, either IVUS/OCT or provocative test is required in the presence of evidence in favor of ischemic origin (Fig. 1).

Fig. 1.

Algorithm showing the work-up for the management of MINOCA. Coronary angiography is immediately recommended in STEMI patients while it could be postponed according to risk stratification in NSTEMI patients. Then, CMRI is recommended in all patients with non-significant CAD followed by endovascular imaging (OCT and IVUS) or provocative test depending on clinical history in the presence of evidence of ischemia. *CAD, coronary artery disease; AMI, acute myocardial infarction; MINOCA, myocardial infarction with non-obstructive coronary artery disease; CMRI, cardiac magnetic resonance imaging; STEMI, ST segment elevation myocardial infarction; NSTEMI, no ST segment elevation myocardial infarction; TTE, transthoracic echocardiography; SCAD, spontaneous coronary dissection.

Excluding non-cardiac disorders that mimickAMI like pulmonary embolism, sepsis and forms of type 2 myocardial infarction (anaemia and hyperthyroidism) is an important step. Thus, detailed clinical history, electrocardiogram, critical laboratory tests (D-dimer, red and white blood cells counts, creatinine, thyroide stimulating hormone) and pulmonary CT-angiography are useful tools depending on clinical presentation. However, Collste et al. [4] did not reported any case of pulmonary embolism after performing CT- angiography in 100 consecutive MINOCA patients. Screening for drug consumption including sympathomimetic agents (cocaine and methamphetamines) and for hypercoagulable state may be helpful in a number of patients [10]. The prevalence of inherited thrombophilia disorders like factor V Leiden, Protein C and S deficiencies detected among MINOCA patients is 14% [10].

Transthoracic echocardiography is an essential non-invasive exam systematically performed in the acute setting to detect AMI-complications, structural heart disease, intra-cardiac thrombus, cardiac tumor (myxoma), tako-tsubo features and wall motion abnormalities [67]. In the setting of MINOCA, interventional cardiologist routinely accomplished coronary angiography with left ventriculogram searching for the angiographic features of apical or mid-ventricular Tako-Tsubo like fisherman’s pot or Hawk’s beak appearance, respectively [68] (Fig. 2).

Fig. 2.

Left ventriculogram and coronary angiography. Left ventriculogram showing mid-ventricular tako-tsubo with hawk’s beak appearance (a) and apical tako-tsubo with fisherman’s pot feature (b) in 2 MINOCA patients. Left coronary angiogram showing patent coronary arteries (c) and severe systolic milking effect of myocardial bridge (d) in MINOCA patient.

Then, cardiac magnetic resonance imaging (C-MRI) is the key diagnostic tool to differentiate ischemic from non-ischemic causes in MINOCA patients. C-MRI is very important in the diagnosis. Subendocardial late godalinium enhancement (Fig. 3a) is most consistent with infarct [3]. Non-ischemic causes are likely mid-wall or sub-epicardial. However, they can be subendocardial as well in cases of sarcoidosis, amyloid. Myocardial edema shows inflammation. That can be present in infarct as well as in non-specific inflammation from many causes of MINOCA [3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23]. Furthermore, C-MRI is the recommended imaging modality of choice to confirm myocardial involvement according to the ESC (European Society of Cardiology) guidelines and it was favourably compared to the gold standard endomyocardial biopsy [52, 48, 69, 70, 71, 72]. It is worthy to mention that C-MRI is normal in a large proportion of MINOCA patients (8–67%), thereby raising the reconsideration for alternative non-cardiac disorders or undetectable small infarct or little widely distributed necrotic myocytes [4, 73, 74, 75, 76, 77, 78, 79]. Performing C-MRI within 7 days from clinical presentation is the optimal time to increase the diagnostic yield [73, 75, 76]. Leurent et al. [73] who did C-MRI at a mean delay of 6.9 days in 107 MINOCA patients were able to find a definitive diagnosis in 90% of cases (60% myocarditis (Fig. 3b), 16% AMI and 14% cardiomyopathy). However, the overall diagnostic yield of C-MRI performed within 4 weeks after hospital admission in MINOCA patients was 71% (33% myocarditis, 12% takotsubo cardiomyopathy, 2% hypertrophic cardiomyopathy, 2% dilated cardiomyopathy and 3% other) [74, 75, 76, 77, 78, 79, 80, 81, 82, 83]. Recently, Dastidar et al. [84] assessed the prognostic role of C-MRI in MINOCA patients and identified C-MRI diagnosis of cardiomyopathy as an independant predictor of mortality.

Fig. 3.

Cardiac magnetic resonance imaging. Short-axis gadolinium enhanced cardiac magnetic resonance imaging showing an acute sub-endocardial necrosis on the infero-septo-basal segment in MINOCA patient with plaque rupture (a) and sub-epicardial enhancement of the lateral wall in MINOCA patient with acute myocarditis (b).

Intracoronary imaging like optical coherence tomography (OCT) and at lesser extent intravascular ultrasound (IVUS) are important tools for the diagnosis of coronary plaque disruption, spontaneous coronary dissection and coronary thrombosis. Plaque disruption including rupture, hemorrhage, ulcer and erosion was found in 40% of MINOCA cases [26]. Documented plaque rupture on OCT was correlated to major adverse cardiac events [85]. Given that plaque erosion is not detected by IVUS, OCT is the preferred intravascular imaging modality [86]. A recent study by Reynold et al. [87] identified a culrpit lesion (plaque rupture) by using OCT in 46.2% of total 145 participating women. They also noticed that multimodality imaging combining C-MRI and OCT were more powerful to determine the underlying cause of MINOCA than each modality alone [87].

Lastly, provocative spasm testing maybe considered in some MINOCA patients who were likely to have vasospastic angina. Recurrent episodes of rest angina with circadian pattern that respond to nitrates are in favor of vasospasm [88]. Inducible major epicardial coronary spasm was revealed in 43 to 54% of MINOCA patients [89, 90] (Fig. 4). Montone et al. [89] recommended intracoronary reactivity testing in all MINOCA patients. He also showed that provocative test could be safely performed in the acute phase following coronary angiography [89, 90]. Currently, Reynolds et al. [87] have identified coronary artery spasm as underlying cause of MINOCA in 46 women over 145 by using OCT. To complete, it is worthy to mention that coronary microvascular spasm (vessels <0.5 mm diameter) was revealed in about 16% to 54% of MINOCA patients [52, 89, 91]. Ischemic ECG changes with chest disconfort after intracoronary injection of acetylcholine without angiographic epicardial coronary artery spasm direct towards microcirculatory spasm [92, 93]. Multiple invasive and non-invasive techniques such as CMRI, doppler transthoracic echocardiography, positron emission tomography and hyperaemic absolute coronary flow measurement are useful to assess coronary microcirculation accounting for 70% of total coronary resistance in the absence of obstructive disease [94, 95].

Fig. 4.

Intracoronary reactivity testing. Intracoronary reactivity testing performed in MINOCA patient: coronary angiogram showed a patent right coronary artery (a) with proximal vasospasm in response to methergin administration (b,c) that resolves after injection of nitroglycerine (d).

4. Therapeutic approach

Since MINOCA involved several pathophysiological mechanisms and various clinical entities, the therapeutic strategy varies depending on the underlying aetiology. However, it remains unclear if treatment strategy for AMI is suitable for MINOCA patients without an identifiable cause. Lifestyle modification including smoking cessation, regular physical activity, weight loss and meditteranean diet was recommended in all MINOCA patients [96]. The beneficial effects of renin-angiotensin converting enzyme inhibitors (ACEI) on reducing mortality and major adverse cardiac events in MINOCA have been reported by previously conducted studies [97, 98, 99] while conflicting results were observed with statins [65, 97, 98, 100]. A neutral effect was linked to antiplatelet therapy which can also be harmful to some MINOCA patients [101, 102].

4.1 Ischemic causes

The management of MINOCA patients with plaque disruption should be in accordance to the guidelines for AMI [7, 103]. Indeed, dual antiplatelet therapy is recommended for 1 year followed by lifelong single antiplatelet therapy. For MINOCA patients with spontaneous coronary artery dissection, a conservative approach is preferred because coronary intervention or stenting contributes to the propagation of the dissection and mural hematoma [104]. β-blockers and aspirin are the mainstay medical therapy while the role of anticoagulant and dual-antiplatelet remains controversial [105, 106]. Some researchers hypothesized that using P2Y12 inhibitors could be beneficial view the prothrombotic state trigerred by the intimal tear [22] while others reported that these agents increased the risk of bleeding and subsequently the progression of intramural hematoma [97]. Statins were not recommended unless atherosclerotic disease is present in SCAD patients [107]. Calcium channel blockers and nitrates were used as first line therapy for the treatment of MINOCA patients with vasospastic angina [90]. Combining dihydropyridine and non dihydropyridine calcium channel blockers or adding nicorandil (potassium channel activator) is indicated for refractory vasospastic angina [88]. Dipyridamole [108] and ranolazine [109] which promote microcirculatory vasodilation and visceral analgesic agents like imipramine [110] and aminophylline [111] are of potential benefits in MINOCA patients with microvascular dysfunction. We highlight on the lack of consensus for secondary prevention implantable cardiac defibrillators in extreme cases of coronary spasmor other causes of MINOCA resulting in sudden cardiac death, although it is likely reasonable.

4.2 Non-ischemic causes

MINOCA patients with myocarditis were conventionally treated with diuretics, ACEI or angiotensin receptor blockade (ARB) and β-blockers [52]. Intravenous inotropic agents or mechanical cardiopulmonary support were required if they are hemodynamically unstable [52]. To date, there are no guidelines on medical treatment for MINOCA with Tako-tsubo cardiomyopathy, and heart failure therapies (β-blockers and ACEI/ARB) were routinely used. Finally, approved medicines for secondary prevention after MI like statins, ACEI or ARB, β-blockers and dual antiplatelent agents were recommended in MINOCA patients with unclear aetiology [93, 112, 113].

5. Prognosis

A recent large nation-wide study showed a comparable mid-term prognosis between MINOCA and CAD-AMI patients [98]. Data from literature are conflicting because while some studies reported a lower mortality rate and cardiovascular events in MINOCA population [98, 114, 115, 116, 117], others revealed similar adverse clinical outcomes between MINOCA and CAD-AMI [118, 119, 120]. Except for revascularization which was more commonly observed among CAD-AMI patients, there are no significant difference in adverse clinical outcomes including death from any cause, cardiac and non-cardiac deaths at 2-years follow-up between MINOCA and CAD-AMI populations [98]. Similarly, the frequencies of in-hospital events were comparable [99]. The use of statins in the presence of mild atherosclerotic disease and ACE/ARB was associated with prolonged survival [2, 91, 92, 93, 94, 98, 113, 114] whereas advanced age, diabetes mellitus, atypical manifestations, STEMI presentation and killip class IV were independent predictors of mortality in MINOCA patients [98, 121].

6. Conclusions

MINOCA is a common clinical entity accounting up to 15% of AMI population and incorporating numerous pathophysiological mechanisms. It sould be considered as a working diagnosis and additional multimodality imaging diagnsotic approach (Left ventriculogam, Echocardiography, C-MRI, IVUS, OCT) is usually needed. Since the therapeutic management differs among MINOCA patients, determining the underlying aetiology is fundamental to perform cause-targeted therapy.

Author contributions

AM contributed to conception, design and writing of the report; VN contributed to conception and design of the report; JR contributed to design and writing of the report and provided important intellectual contributions to the manuscript.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

We would like to express our gratitude to all the peer reviewers for their opinions and suggestions during the writing of this manuscript.

Funding

This research received no external funding.

Conflict of interest

The authors declare no conflict of interest.

References
[1]
Bainey KR, Welsh RC, Alemayehu W, Westerhout CM, Traboulsi D, Anderson T, et al. Population-level incidence and outcomes of myocardial infarction with non-obstructive coronary arteries (MINOCA): Insights from the Alberta contemporary acute coronary syndrome patients invasive treatment strategies (COAPT) study. International Journal of Cardiology. 2018; 264: 12–17.
[2]
Safdar B, Spatz ES, Dreyer RP, Beltrame JF, Lichtman JH, Spertus JA, et al. Presentation, Clinical Profile, and Prognosis of Young Patients with Myocardial Infarction with Nonobstructive Coronary Arteries (MINOCA): Results from the VIRGO Study. Journal of the American Heart Association. 2018; 7: e009174.
[3]
Bairey Merz CN, Pepine CJ, Walsh MN, Fleg JL. Ischemia and no Obstructive Coronary Artery Disease (INOCA): Developing Evidence-Based Therapies and Research Agenda for the next Decade. Circulation. 2017; 135: 1075–1092.
[4]
Collste O, Sörensson P, Frick M, Agewall S, Daniel M, Henareh L, et al. Myocardial infarction with normal coronary arteries is common and associated with normal findings on cardiovascular magnetic resonance imaging: results from the Stockholm Myocardial Infarction with Normal Coronaries study. Journal of Internal Medicine. 2013; 273: 189–196.
[5]
Pasupathy S, Tavella R, Beltrame JF. Myocardial Infarction with Nonobstructive Coronary Arteries (MINOCA): the Past, Present, and Future Management. Circulation. 2017; 135: 1490–1493.
[6]
Agewall S, Beltrame JF, Reynolds HR, Niessner A, Rosano G, Caforio ALP, et al. ESC working group position paper on myocardial infarction with non-obstructive coronary arteries. European Heart Journal. 2017; 38: 143–153.
[7]
Thygesen K, Alpert JS, Jaffe AS, Chaitman BR, Bax JJ, Morrow DA, et al. Fourth Universal Definition of Myocardial Infarction (2018). Circulation. 2018; 138: e618–e651.
[8]
Barr PR, Harrison W, Smyth D, Flynn C, Lee M, Kerr AJ. Myocardial Infarction without Obstructive Coronary Artery Disease is not a Benign Condition (ANZACS-QI 10). Heart, Lung & Circulation. 2018; 27: 165–174.
[9]
Daniel M, Agewall S, Caidahl K, Collste O, Ekenbäck C, Frick M, et al. Effect of Myocardial Infarction with Nonobstructive Coronary Arteries on Physical Capacity and Quality-of-Life. The American Journal of Cardiology. 2017; 120: 341–346.
[10]
Pasupathy S, Air T, Dreyer RP, Tavella R, Beltrame JF. Systematic review of patients presenting with suspected myocardial infarction and nonobstructive coronary arteries. Circulation. 2015; 131: 861–870.
[11]
Pelliccia F, Pasceri V, Niccoli G, Tanzilli G, Speciale G, Gaudio C, et al. Predictors of Mortality in Myocardial Infarction and Nonobstructed Coronary Arteries: a Systematic Review and Meta-Regression. The American Journal of Medicine. 2020; 133: 73–83.e4.
[12]
Abdu FA, Liu L, Mohammed A, Luo Y, Xu S, Auckle R, et al. Myocardial infarction with non-obstructive coronary arteries (MINOCA) in Chinese patients: Clinical features, treatment and 1 year follow-up. International Journal of Cardiology. 2019; 287: 27–31.
[13]
Nordenskjöld AM, Baron T, Eggers KM, Jernberg T, Lindahl B. Predictors of adverse outcome in patients with myocardial infarction with non-obstructive coronary artery (MINOCA) disease. International Journal of Cardiology. 2018; 261: 18–23.
[14]
Ciliberti G, Coiro S, Tritto I, Benedetti M, GuerraF, Del Pinto M, et al. Predictors of poor clinical outcomes in patients with acute myocardial infarction and non-obstructed coronary arteries (MINOCA). International Journal of Cardiology. 2018; 267: 41–45.
[15]
Hjort M, Lindahl B, Baron T, Jernberg T, Tornvall P, Eggers KM. Prognosis in relation to high-sensitivity cardiac troponin T levels in patients with myocardial infarction and non-obstructive coronary arteries. American Heart Journal. 2018; 200: 60–66.
[16]
Planer D, Mehran R, Ohman EM, White HD, Newman JD, Xu K, et al. Prognosis of patients with non-ST-segment-elevation myocardial infarction and nonobstructive coronary artery disease: propensity-matched analysis from the Acute Catheterization and Urgent Intervention Triage Strategy trial. Circulation. Cardiovascular Interventions. 2014; 7: 285–293.
[17]
Montenegro Sá F, Ruivo C, Santos LG, Antunes A, Saraiva F, Soares F, et al. Myocardial infarction with nonobstructive coronary arteries: a single-center retrospective study. Coronary Artery Disease. 2018; 29: 511–515.
[18]
Alfonso F, Paulo M, Dutary J. Endovascular Imaging of Angiographically Invisible Spontaneous Coronary Artery Dissection. JACC: Cardiovascular Interventions. 2012; 5: 452–453.
[19]
Antonsen L, Thayssen P, Jensen LO. Large coronary intramural hematomas: a case series and focused literature review. Cardiovascular Revascularization Medicine. 2015; 16: 116–123.
[20]
Saw J, Aymong E, Sedlak T, Buller CE, Starovoytov A, Ricci D, et al. Spontaneous coronary artery dissection: association with predisposing arteriopathies and precipitating stressors and cardiovascular outcomes. Circulation: Cardiovascular Interventions. 2014; 7: 645–655.
[21]
Saw J, Aymong E, Mancini GBJ, Sedlak T, Starovoytov A, Ricci D. Nonatherosclerotic coronary artery disease in young women. The Canadian Journal of Cardiology. 2014; 30: 814–819.
[22]
Saw J, Mancini GBJ, Humphries KH. Contemporary Review on Spontaneous Coronary Artery Dissection. Journal of the American College of Cardiology. 2016; 68: 297–312.
[23]
Tweet MS, Gulati R, Hayes SN. Spontaneous Coronary Artery Dissection. Current Cardiology Reports. 2016; 18: 60.
[24]
Jackson R, Al-Hussaini A, Joseph S, van Soest G, Wood A, Macaya F, et al. Spontaneous Coronary Artery Dissection. JACC: Cardiovascular Imaging. 2019; 12: 2475–2488.
[25]
Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD, et al. Third universal definition of myocardial infarction. European Heart Journal. 2012; 33: 2551–2567.
[26]
Reynolds HR, Srichai MB, Iqbal SN, Slater JN, Mancini GBJ, Feit F, et al. Mechanisms of myocardial infarction in women without angiographically obstructive coronary artery disease. Circulation. 2011; 124: 1414–1425.
[27]
Ouldzein H, Elbaz M, Roncalli J, Cagnac R, Carrié D, Puel J, et al. Plaque rupture and morphological characteristics of the culprit lesion in acute coronary syndromes without significant angiographic lesion: analysis by intravascular ultrasound. Annales de Cardiologie et d’Angiologie. 2011; 61: 20–26.
[28]
Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arteriosclerosis, Thrombosis, and Vascular Biology. 2000; 20: 1262–1275.
[29]
Mittleman MA, Mostofsky E. Physical, psychological and chemical triggers of acute cardiovascular events: preventive strategies. Circulation. 2011; 124: 346–354.
[30]
Hao H, Gabbiani G, Camenzind E, Bacchetta M, Virmani R, Bochaton-Piallat M. Phenotypic Modulation of Intima and Media Smooth Muscle Cells in Fatal Cases of Coronary Artery Lesion. Arteriosclerosis, Thrombosis, and Vascular Biology. 2006; 26: 326–332.
[31]
Kaski JC, Crea F, Meran D, Rodriguez L, Araujo L, Chierchia S, et al. Local coronary supersensitivity to diverse vasoconstrictive stimuli in patients with variant angina. Circulation. 1986; 74: 1255–1265.
[32]
Pristipino C, Beltrame JF, Finocchiaro ML, Hattori R, Fujita M, Mongiardo R, et al. Major racial differences in coronary constrictor response between japanese and caucasians with recent myocardial infarction. Circulation. 2000; 101: 1102–1108.
[33]
Smith KJ, Chadburn AJ, Adomaviciene A, Minoretti P, Vignali L, Emanuele E, et al. Coronary spasm and acute myocardial infarction due to a mutation (V734I) in the nucleotide binding domain 1 of ABCC9. International Journal of Cardiology. 2013; 168: 3506–3513.
[34]
Mohri M, Koyanagi M, Egashira K, Tagawa H, Ichiki T, Shimokawa H, et al. Angina pectoris caused by coronary microvascular spasm. Lancet. 1998; 351: 1165–1169.
[35]
Lanza GA, Crea F. Primary coronary microvascular dysfunction: clinical presentation, pathophysiology, and management. Circulation. 2010; 121: 2317–2325.
[36]
Arrebola-Moreno AL, Arrebola JP, Moral-Ruiz A, Ramirez-Hernandez JA, Melgares-Moreno R, Kaski JC. Coronary microvascular spasm triggers transient ischemic left ventricular diastolic abnormalities in patients with chest pain and angiographically normal coronary arteries. Atherosclerosis. 2014; 236: 207–214.
[37]
Crea F, Bairey Merz CN, Beltrame JF, Kaski JC, Ogawa H, Ong P, et al. The parallel tales of microvascular angina and heart failure with preserved ejection fraction: a paradigm shift. European Heart Journal. 2017; 38: 473–477.
[38]
Cuculi F, Togni M, Meier B. Myocardial infarction due to paradoxical embolism in a patient with large atrial septal defect. Journal of Invasive Cardiology. 2009; 21: 84–186.
[39]
Crump R, Shanding AH, Van Natta B, Ellestad M. Prevalence of patent formaen ovale in patients with acute myocardial infarction and angiographically normal coronary arteries. American Journal of Cardiology. 2000; 85: 1368–1370.
[40]
Sastry S, Riding G, Morris J, Taberner D, Cherry N, Heagerty A, et al. Young Adult Myocardial Infarction and Ischemic Stroke: the role of paradoxical embolism and thrombophilia (the YAMIS Study). Journal of the American College of Cardiology. 2006; 48: 686–691.
[41]
Canty JM. Coronary Blood Flow and Myocardial Ischemia. Braunwald’s Heart Disease. 2012; 29: 1049–1075.
[42]
Braunwald E. 50th anniversary historical article. Myocardial oxygen consumption: the quest for its determinants and some clinical fallout. Journal of the American College of Cardiology. 1999; 34: 1365–1368.
[43]
Rhee JW, Sabatine MS, Lilly LS. Ischemic heart disease. Pathophysiology of Heart Disease: a collaborative project of medical students and faculty. Philadelphia, PA: Lipincott Williams & Wilkins. 2011; 135–160.
[44]
Tornvall P, Gerbaud E, Behaghel A, Chopard R, Collste O, Laraudogoitia E, et al. Myocarditis or “true” infarction by cardiac magnetic resonance in patients with a clinical diagnosis of myocardial infarction without obstructive coronary disease: A meta-analysis of individual patient data. Atherosclerosis. 2015; 241: 87–91.
[45]
Mahrholdt H, Wagner A, Deluigi CC, Kispert E, Hager S, Meinhardt G, et al. Presentation, patterns of myocardial damage, and clinical course of viral myocarditis. Circulation. 2006; 114: 1581–1590.
[46]
Kühl U, Pauschinger M, Bock T, Klingel K, Schwimmbeck CPL, Seeberg B, et al. Parvovirus B19 infection mimicking acute myocardial infarction. Circulation. 2003; 108: 945–950.
[47]
Yilmaz A, Mahrholdt H, Athanasiadis A, Vogelsberg H, Meinhardt G, Voehringer M, et al. Coronary vasospasm as the underlying cause for chest pain in patients with PVB19 myocarditis. Heart. 2008; 94: 1456–1463.
[48]
Leone O, Veinot JP, Angelini A, Baandrup UT, Basso C, Berry G, et al. 2011 consensus statement on endomyocardial biopsy from the Association for European Cardiovascular Pathology and the Society for Cardiovascular Pathology. Cardiovascular Pathology. 2012; 21: 245–274.
[49]
Caforio ALP, Marcolongo R, Jahns R, Fu M, Felix SB, Iliceto S. Immune-mediated and autoimmune myocarditis: clinical presentation, diagnosis and management. Heart Failure Reviews. 2013; 18: 715–732.
[50]
Bock C, Klingel K, Kandolf R. Human parvovirus B19-associated myocarditis. The New England Journal of Medicine. 2010; 362: 1248–1249.
[51]
Caforio AL, Keeling PJ, Zachara E, Mestroni L, Camerini F, Mann JM, et al. Evidence from family studies for autoimmunity in dilated cardiomyopathy. Lancet. 1994; 344: 773–777.
[52]
Caforio ALP, Pankuweit S, Arbustini E, Basso C, Gimeno-Blanes J, Felix SB, et al. Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. European Heart Journal. 2013; 34: 2636–2648.
[53]
Galiuto L, De Caterina AR, Porfidia A, Paraggio L, Barchetta S, Locorotondo G, et al. Reversible coronary microvascular dysfunction: a common pathogenic mechanism in apical ballooning or takotsubo syndrome. European Heart Journal. 2010; 31: 1319–1327.
[54]
Manolis AS, Manolis AA, Manolis TA, Melita H. COVID-19 and Acute Myocardial Injury and Infarction: Related Mechanisms and Emerging Challenges. Journal of Cardiovascular Pharmacology and Therapeutics. 2021; 26: 399–414.
[55]
Joly BS, Siguret V, Veyradier A. Understanding pathophysiology of hemostasis disorders in critically ill patients with COVID-19. Intensive Care Medicine. 2020; 46: 1603–1606.
[56]
Lodigiani C, Iapichino G, Carenzo L, Cecconi M, Ferrazzi P, Sebastian T, et al. Humanitas COVID-19 Task Force Venous and Arterial Thromboembolic Complications in COVID-19 Patients Admitted to an Academic Hospital in Milan, Italy. Journal of Vascular Surgery: Venous and Lymphatic Disorders. 2020; 8: 1119.
[57]
Spiezia L, Boscolo A, Poletto F, Cerruti L, Tiberio I, Campello E, et al. COVID-19-Related Severe Hypercoagulability in Patients Admitted to Intensive Care Unit for Acute Respiratory Failure. Thrombosis and Haemostasis. 2020; 120: 998–1000.
[58]
Matta A, Canitrot R, Nader V, Blanco S, Campelo-Parada F, Bouisset F, et al. Left anterior descending myocardial bridge: Angiographic prevalence and its association to atherosclerosis. Indian Heart Journal. 2021. (in press)
[59]
Ishikawa Y, Akasaka Y, Suzuki K, Fujiwara M, Ogawa T, Yamazaki K, et al. Anatomic Properties of Myocardial Bridge Predisposing to Myocardial Infarction. Circulation. 2009; 120: 376–383.
[60]
Ge J, Huang Z, Liu X, Qian J. Spontaneous coronary dissection associated with myocardial bridge causing acute myocardial infarction. Chinese Medical Journal. 2008; 121: 2450–2453.
[61]
Kim JW, Seo HS, Na JO, Suh SY, Choi CU, Kim EJ, et al. Myocardial bridging is related to endothelial dysfunction but not to plaque as assessed by intracoronary ultrasound. Heart. 2008; 94: 765–769.
[62]
Farag A, Al-Najjar Y, Eichhöfer J. Adenosine-induced vasospasticity in a myocardial bridge…endothelial dysfunction? JACC. Cardiovascular Interventions. 2015; 8: e21–e22.
[63]
Duygu H. A bridge to coronary spasm: Myocardial bridging. International Journal of Cardiology. 2016; 214: 35–36.
[64]
Mookadam F, Green J, Holmes D, Moustafa SE, Rihal C. Clinical relevance of myocardial bridging severity: single center experience. European Journal of Clinical Investigation. 2009; 39: 110–115.
[65]
Hong L, Liu J, Luo S, Li J. Relation of myocardial bridge to myocardial infarction: a meta-analysis. Chinese Medical Journal. 2014; 127: 945–950.
[66]
Rakowski T, De Luca G, Siudak Z, Plens K, Dziewierz A, Kleczyński P, et al. Characteristics of patients presenting with myocardial infarction with non-obstructive coronary arteries (MINOCA) in Poland: data from the ORPKI national registry. Journal of Thrombosis and Thrombolysis. 2019; 47: 462–466.
[67]
Pustjens TFS, Appelman Y, Damman P, ten Berg JM, Jukema JW, de Winter RJ, et al. Guidelines for the management of myocardial infarction/injury with non-obstructive coronary arteries (MINOCA): a position paper from the Dutch ACS working group. Netherlands Heart Journal. 2020; 28: 116–130.
[68]
Matta A, Roncalli J, Elbaz M, Lhermusier T, Campelo-Parada F, Bouisset F, et al. Mid-ventricular Takotsubo cardiomyopathy with hawk’s beak appearance: a case report. American Journal of Case Reports. 2020; 21: e919563.
[69]
Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, et al. Heart Failure and Transplantation Committee, Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups, Council on Epidemiology and Prevention. Contemporary definitions and classifications of cardiomyopathies. An American Heart Association Scientific statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee, quality of Care and Outcome research and Functional Genomics and Translational Biology Interdisciplinary Working Groups: and Council on Epidemiology and Prevention. Circulation. 2006; 113: 1807–1816.
[70]
Elliott P, Andersson B, Arbustini E, Bilinska Z, Cecchi F, Charron P, et al. Classification of the cardiomyopathies: a position statement from the European Society of Cardiology working group on myocardial and pericardial disease. European Heart Journal. 2008; 29: 270–276.
[71]
Lurz P, Eitel I, Adam J, Steiner J, Grothoff M, Desch S, et al. Diagnostic Performance of CMR Imaging Compared with EMB in Patients with Suspected Myocarditis. JACC: Cardiovascular Imaging. 2012; 5: 513–524.
[72]
Adler Y, Charron P, Imazio M, Badano L, Barón-Esquivias G, Bogaert J, et al. 2015 ESC Guidelines for the diagnosis and management of pericardial diseases: the Task Force for the Diagnosis and Management of Pericardial Diseases of the European Society of Cardiology (ESC)Endorsed by: the European Association for Cardio-Thoracic Surgery (EACTS). European Heart Journal. 2015; 36: 2921–2964.
[73]
Leurent G, Langella B, Fougerou C, Lentz P, Larralde A, Bedossa M, et al. Diagnostic contributions of cardiac magnetic resonance imaging in patients presenting with elevated troponin, acute chest pain syndrome and unobstructed coronary arteries. Archives of Cardiovascular Diseases. 2011; 104: 161–170.
[74]
Stensaeth KH, Fossum E, Hoffmann P, Mangschau A, Klow NE. Clinical characteristics and role of early cardiac magnetic resonance imaging in patients with suspected ST-elevation myocardial infarction and normal coronary arteries. The International Journal of Cardiovascular Imaging. 2011; 27: 355–365.
[75]
Baccouche H, Mahrholdt H, Meinhardt G, Merher R, Voehringer M, Hill S, et al. Diagnostic synergy of non-invasive cardiovascular magnetic resonance and invasive endomyocardial biopsy in troponin-positive patients without coronary artery disease. European Heart Journal. 2009; 30: 2869–2879.
[76]
Gerbaud E, Harcaut E, Coste P, Erickson M, Lederlin M, Labèque JN, et al. Cardiac magnetic resonance imaging for the diagnosis of patients presenting with chest pain, raised troponin, and unobstructed coronary arteries. The International Journal of Cardiovascular Imaging. 2012; 28: 783–794.
[77]
Avegliano GP, Huguet M, Costabel JP, Kuschnir P, Thierer J, Alves de Lima A, et al. Cardiac magnetic resonance imaging in patients with chest pain, high troponin levels and absence of coronary artery obstruction. Revista Argentina de Cardiologia. 2011; 79: 226–230.
[78]
Zaldumbide EL, Pérez-David E, Larena JA, del Castillo SV, Rumoroso Cuevas JR, Onaindía JJ, et al. The Value of Cardiac Magnetic Resonance in Patients with Acute Coronary Syndrome and Normal Coronary Arteries. Revista EspañOla De Cardiología. 2009; 62: 976–983.
[79]
Eitel I, Behrendt F, Schindler K, Kivelitz D, Gutberlet M, Schuler G, et al. Differential diagnosis of suspected apical ballooning syndrome using contrast-enhanced magnetic resonance imaging. European Heart Journal. 2008; 29: 2651–2659.
[80]
Alsaileek A, Nasim M, Aljizeeri A, Alharthi M, Al-Mallah MH. The role of delayed contrast-enhanced cardiac magnetic resonance in differentiating myocarditis from myocardial infarction. European Heart Journal Supplements. 2014; 16: B24–B28.
[81]
Chopard R, Jehl J, Dutheil J, Genon VD, Seronde M, Kastler B, et al. Evolution of acute coronary syndrome with normal coronary arteries and normal cardiac magnetic resonance imaging. Archives of Cardiovascular Diseases. 2011; 104: 509–517.
[82]
Assomull RG, Lyne JC, Keenan N, Gulati A, Bunce NH, Davies SW, et al. The role of cardiovascular magnetic resonance in patients presenting with chest pain, raised troponin, and unobstructed coronary arteries. European Heart Journal. 2007; 28: 1242–1249.
[83]
Mahmoudi M, Harden S, Abid N, Peebles C, Nicholas Z, Jones T, et al. Troponin-positive chest pain with unobstructed coronary arteries: definitive differential diagnosis using cardiac MRI. The British Journal of Radiology. 2012; 85: e461–e466.
[84]
Dastidar AG, Baritussio A, De Garate E, Drobni Z, Biglino G, Singhal P, et al. Prognostic role of cardiac MRI and conventional risk factors in myocardial infarction with nonobstructed coronary arteries. Journal of the American College of Cardiology: Cardiovascular Imaging. 2019; 12: 1973–1982.
[85]
Niccoli G, Scalone G, Crea F. Acute myocardial infarction with no obstructive coronary atherosclerosis: mechanisms and management. European Heart Journal. 2015; 36: 475–481.
[86]
Kubo T, Imanishi T, Takarada S, Kuroi A, Ueno S, Yamano T, et al. Assessment of culprit lesion morphology in acute myocardial infarction: ability of optical coherence tomography compared with intravascular ultrasound and coronary angioscopy. Journal of the American College of Cardiology. 2007; 50: 933–939.
[87]
Reynolds HR, Maehara A, Kwong RY, Sedlak T, Saw J, Smilowitz NR, et al. Coronary Optical Coherence Tomography and Cardiac Magnetic Resonance Imaging to Determine Underlying Causes of Myocardial Infarction with Nonobstructive Coronary Arteries in Women. Circulation. 2021; 143: 624–640.
[88]
Matta A, Bouisset F, Lhermusier T, Campelo-Parada F, Elbaz M, Carrié D, et al. Coronary Artery Spasm: New Insights. Journal of Interventional Cardiology. 2020; 2020: 1–10.
[89]
Montone RA, Niccoli G, Russo M, Giaccari M, Del Buono MG, Meucci MC, et al. Clinical, angiographic and echocardiographic correlates of epicardial and microvascular spasm in patients with myocardial ischaemia and non-obstructive coronary arteries. Clinical Research in Cardiology. 2020; 109: 435–443.
[90]
Pirozzolo G, Seitz A, Athanasiadis A, Bekeredjian R, Sechtem U, Ong P. Microvascular spasm in non-ST-segment elevation myocardial infarction without culprit lesion (MINOCA). Clinical Research in Cardiology. 2020; 109: 246–254.
[91]
Da Costa A, Isaaz K, Faure E, Mourot S, Cerisier A, Lamaud M. Clinical characteristics, aetiologic factors and long-term prognosis of myocardial infarction with an absolutely normal coronary angiogram: a 3-year follow-up study of 91 patients. European Heart Journal. 2001; 22: 1459–1465.
[92]
Ong P, Athanasiadis A, Borgulya G, Vokshi I, Bastiaenen R, Kubik S, et al. Clinical usefulness, angiographic characteristics, and safety evaluation of intracoronary acetylcholine provocation testing among 921 consecutive white patients with unobstructed coronary arteries. Circulation. 2014; 129: 1723–1730.
[93]
Tamis-Holland JE, Jneid H, Reynolds HR, Agewall S, Brilakis ES, Brown TM, et al. Contemporary Diagnosis and Management of Patients with Myocardial Infarction in the Absence of Obstructive Coronary Artery Disease: a Scientific Statement from the American Heart Association. Circulation. 2019; 139: 891–908.
[94]
Xaplanteris P, Fournier S, Keulards DCJ, Adjedj J, Ciccarelli G, Milkas A, et al. Catheter-Based Measurements of Absolute Coronary Blood Flow and Microvascular Resistance: Feasibility, Safety, and Reproducibility in Humans. Circulation. Cardiovascular Interventions. 2018; 11: e006194.
[95]
Beltrame JF, Crea F, Camici P. Advances in Coronary Microvascular Dysfunction. Heart, Lung and Circulation. 2009; 18: 19–27.
[96]
Pacheco Claudio C, Quesada O, Pepine CJ, Noel Bairey Merz C. Why names matter for women: MINOCA/INOCA (myocardial infarction/ischemia and no obstructive coronary artery disease). Clinical Cardiology. 2018; 41: 185–193.
[97]
Lindahl B, Baron T, Erlinge D, Hadziosmanovic N, Nordenskjöld A, Gard A, et al. Medical Therapy for Secondary Prevention and Long-Term Outcome in Patients with Myocardial Infarction with Nonobstructive Coronary Artery Disease. Circulation. 2017; 135: 1481–1489.
[98]
Choo EH, Chang K, Lee KY, Lee D, Kim JG, Ahn Y, et al. Prognosis and Predictors of Mortality in Patients Suffering Myocardial Infarction with Non‐Obstructive Coronary Arteries. Journal of the American Heart Association. 2019; 8: e011990.
[99]
Cenko E, Manfrini O, Morell C, Das R, Barth JH, Hall AS, et al. Angiotensin-converting enzyme inhibitor therapy in patients with non-obstructive coronary artery disease. European Heart Journal. 2013; 34: 2278.
[100]
Cespon Fernandez M, Abu-Assi E, Raposeiras-Roubin S, Munoz-Pousa I, Dominguez-Rodriguez LM, Dominguez-Erquicia P, et al. Cardiovascular mortality in patients with MINOCA and prognostic effect of statin treatment. European Heart Journal. 2019;
[101]
Ishii M, Kaikita K, Sato K, Yamanaga K, Miyazaki T, Akasaka T, et al. Impact of aspirin on the prognosis in patients with coronary spasm without significant atherosclerotic stenosis. International Journal of Cardiology. 2016; 220: 328–332.
[102]
Bossard M, Yusuf S, Tanguay JF, Faxon DP, Boden WE, Steg PG, et al. Recurrent cardiovascular events and mortality in relation to antiplatelet therapy in patients with myocardial infarction without obstructive coronary artery disease (MINOCA). European Heart Journal. 2019; 40.
[103]
Lindahl B, Baron T, Erlinge D, Hadziosmanovic N, Nordenskjold A, Gard A, et al. Medical therapy for secondary prevention and long-term outcome in patients with nonobstructive coronary artery disease. Circulation. 2017; 135: 1481–1489.
[104]
Tweet MS, Eleid MF, Best PJM, Lennon RJ, Lerman A, Rihal CS, et al. Spontaneous coronary artery dissection: revascularization versus conservative therapy. Circulation. Cardiovascular Interventions. 2014; 7: 777–786.
[105]
Mahmoud AN, Taduru SS, Mentias A, Mahtta D, Barakat AF, Saad M, et al. Trends of Incidence, Clinical Presentation, and in-Hospital Mortality among Women with Acute Myocardial Infarction with or without Spontaneous Coronary Artery Dissection. JACC: Cardiovascular Interventions. 2018; 11: 80–90.
[106]
Adlam D, Alfonso F, Maas A, Vrints C, al-Hussaini A, Bueno H, et al. European Society of Cardiology, acute cardiovascular care association, SCAD study group: a position paper on spontaneous coronary artery dissection. European Heart Journal. 2018; 39: 3353–3368.
[107]
Tweet MS, Hayes SN, Pitta SR, Simari RD, Lerman A, Lennon RJ, et al. Clinical features, management, and prognosis of spontaneous coronary artery dissection. Circulation. 2012; 126: 579–588.
[108]
Kurtoglu N, Akcay A, Dindar I. Usefulness of oral dipyridamole therapy for angiographic slow coronary artery flow. The American Journal of Cardiology. 2001; 87: 777–779.
[109]
Saha S, Ete T, Kapoor M, Jha PK, Megeji RD, Kavi G, et al. Effect of Ranolazine in Patients with Chest Pain and Normal Coronaries- a Hospital Based Study. Journal of Clinical and Diagnostic Research. 2017; 11: OC14–OC16.
[110]
Cannon RO, Quyyumi AA, Mincemoyer R, Stine AM, Gracely RH, Smith WB, et al. Imipramine in patients with chest pain despite normal coronary angiograms. The New England Journal of Medicine. 1994; 330: 1411–1417.
[111]
Elliott PM, Krzyzowska-Dickinson K, Calvino R, Hann C, Kaski JC. Effect of oral aminophylline in patients with angina and normal coronary arteriograms (cardiac syndrome X). Heart. 1997; 77: 523–526.
[112]
Pasupathy S, Tavella R, Beltrame JF. The what, when, who, why, how and where of Myocardial Infarction with Non-Obstructive Coronary Arteries (MINOCA). Circulation Journal. 2016; 80: 11–16.
[113]
Manfrini O, Morrell C, Das R, Barth JH, Hall AS, Gale CP, et al. Effects of angiotensin-converting enzyme inhibitors and beta blockers on clinical outcomes in patients with and without coronary artery obstructions at angiography (from a Register-Based Cohort Study on Acute Coronary Syndromes). The American Journal of Cardiology. 2014; 113: 1628–1633.
[114]
Roe MT, Harrington RA, Prosper DM, Pieper KS, Bhatt DL, Lincoff AM, et al. Clinical and therapeutic profile of patients presenting with acute coronary syndromes who do not have significant coronary artery disease. The Platelet Glycoprotein IIb/IIIa in Unstable Angina: Receptor Suppression Using Integrilin Therapy (PURSUIT) Trial Investigators. Circulation. 2000; 102: 1101–1106.
[115]
Diver DJ, Bier JD, Ferreira PE, Sharaf BL, McCabe C, Thompson B, et al. Clinical and arteriographic characterization of patients with unstable angina without critical coronary arterial narrowing (from the TIMI-IIIA Trial). The American Journal of Cardiology. 1994; 74: 531–537.
[116]
Patel MR, Chen AY, Peterson ED, Newby LK, Pollack CV, Brindis RG, et al. Prevalence, predictors, and outcomes of patients with non-ST-segment elevation myocardial infarction and insignificant coronary artery disease: results from the can Rapid risk stratification of Unstable angina patients Suppress ADverse outcomes with Early implementation of the ACC/AHA Guidelines (CRUSADE) initiative. American Heart Journal. 2006; 152: 641–647.
[117]
Pizzi C, Xhyheri B, Costa GM, Faustino M, Flacco ME, Gualano MR, et al. Nonobstructive Versus Obstructive Coronary Artery Disease in Acute Coronary Syndrome: a Meta-Analysis. Journal of the American Heart Association. 2016; 5: e004185.
[118]
Dokainish H, Pillai M, Murphy SA, DiBattiste PM, Schweiger MJ, Lotfi A, et al. Prognostic implications of elevated troponin in patients with suspected acute coronary syndrome but no critical epicardial coronary disease: a TACTICS-TIMI-18 substudy. Journal of the American College of Cardiology. 2005; 45: 19–24.
[119]
Andersson HB, Pedersen F, Engstrøm T, Helqvist S, Jensen MK, Jørgensen E, et al. Long-term survival and causes of death in patients with ST-elevation acute coronary syndrome without obstructive coronary artery disease. European Heart Journal. 2018; 39: 102–110.
[120]
Redondo-Diéguez A, Gonzalez-Ferreiro R, Abu-Assi E, Raposeiras-Roubin S, Aidhodjayeva O, López-López A, et al. Long-term Prognosis of Patients with Non—ST-segment Elevation Acute Myocardial Infarction and Coronary Arteries without Significant Stenosis. Revista EspañOla De Cardiología. 2015; 68: 777–784.
[121]
Fujino M, Ishihara M, Ogawa H, Nakao K, Yasuda S, Noguchi T, et al. Impact of symptom presentation on in‐hospital outcomes in patients with acute myocardial infarction. Journal of Cardiology. 2017; 70: 29–34.
Share
Back to top