Next Article in Journal
It Didn’t Go Away: The Political and Social Determinants of COVID-19 Mortality Rates across Counties in the United States
Previous Article in Journal
Infection Prevention and Control in Public Hospitals and COVID-19 Temporary Treatment and Monitoring Facilities in the Philippines: Results of a Baseline Survey
Previous Article in Special Issue
Quantifying the Impact of COVID-19 “Lockdown” on Physical Activity in Children and Adults with Implanted Cardiac Rhythm Devices: A Single Center Experience Using Cardiac Rhythm Device Accelerometer Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

A Scoping Review on COVID-19-Induced Cardiovascular Complications

1
Department of Pharmacy Practice, ISF College of Pharmacy, Moga 142001, Punjab, India
2
Department of Pharmacology, Central University of Punjab, Ghudda 151401, Punjab, India
*
Author to whom correspondence should be addressed.
COVID 2023, 3(3), 348-369; https://doi.org/10.3390/covid3030026
Submission received: 3 February 2023 / Revised: 1 March 2023 / Accepted: 2 March 2023 / Published: 3 March 2023

Abstract

:
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a type of human coronavirus that resulted in the 2019 coronavirus disease (COVID-19). Although it was generally categorized as a respiratory disease, its involvement in cardiovascular complications was identified from the onset. Elevated cardiac troponin levels (a myocardial injury marker) and echocardiograms, which showed the anomalous performance of the patients’ hearts, were noted in the early case reports obtained from Wuhan, China. A couple of mechanisms have been proposed to explain COVID-19-induced cardiovascular complications, with systemic inflammation being the major focus recently. Chest pain and palpitations are among the prevalent symptoms in moderate to severe COVID-19-recovering patients. Cardiac damage potentially occurs due to multifactorial factors, which include cytokine-induced inflammation, direct cardiotoxicity, and disseminated intravascular coagulation (DIC), among others. The cardiovascular manifestations include cardiac arrhythmia, cardiogenic shock, venous thromboembolism, and elevated cardiac biomarkers. Both the long- and short-term effects of these cardiovascular complications remain puzzling to researchers, as substantial evidence is yet to be gathered to reach a consensus on the severity of COVID-19 in the heart. The treatment considerations currently include antiarrhythmic management, ACEI or ARB use, anticoagulation, hemodynamic support, and immunosuppression. This review aimed to outline the pathogenesis of the various cardiac complications due to COVID-19 as well as the available treatment modalities of COVID-19 infection. Both the mechanisms and the treatments have been succinctly explained in a proper manner to ensure understanding.

1. Introduction

As of 22 August 2022, Turkmenistan remains the only country without any COVID-19 reported cases, according to WHO reports. This is exceptional, especially because it is already about 2.5 years since the COVID-19 pandemic was declared, and currently, there is a total of about 591 million confirmed cases globally, with the United States of America leading, recording the highest number of cases. COVID-19 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and it was first reported to the WHO as pneumonia of unknown cause on 31 December 2019 from Wuhan city, China [1]. SARS-CoV-2 is a single-stranded and RNA-enveloped virus [2]. Its resultant infection includes symptoms such as fatigue, dry cough, fever, and anorexia [3,4]. The family of SARS-CoV viruses has been identified to infect humans earlier on, and SARS-CoV-2 is the seventh known human coronavirus [5].
Cardiovascular complications in COVID-19 patients have been common, and they are among the highest comorbidities in COVID-19 patients. In a study conducted by the Chinese Centre for Disease Control and Prevention, about 44,672 laboratory-confirmed cases were evaluated, and 4.7% of cases were classified as critically ill (multiorgan failure, septic shock, and/or respiratory failure), having the potential of advancing to acute respiratory distress syndrome (ARDS). Higher mortality and more severe cardiovascular complications were observed in COVID-19 patients with cardiovascular comorbidities [6,7]. Moreover, the development of cardiovascular complications during COVID-19 was noted in patients without a CVD history, which could ultimately result in detrimental outcomes during the infection. Cardiac biomarkers elevation (which is used to predict mortality), as well as vascular injury, may be caused by COVID-19, hence the need to understand the mechanisms behind these clinical manifestations [8,9]. In the most critically ill patients, CVD was found to be among the major cause of death in COVID-19 patients [10,11,12].

2. Materials and Methods

A rapid evidence review was conducted, and the scientific literature used mainly constituted original articles sourced from online databases like (PubMed, Cochrane, Web of Sciences, and Google Scholar). Then a careful screening based on the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA-P) was conducted so as to compound the wide pool of information collected, as shown in Figure 1 below. Keywords such as “COVID-19, Cardiovascular complications, recent therapy in COVID-19 treatment, mechanisms of COVID-19” were used.

3. Mechanisms of Cardiovascular Complications in COVID-19

3.1. ACE-2 Inhibitors Downregulation

There are still divergent perspectives on the COVID-19 pathophysiology; however, prior epidemics (SARS and MERS) have been of great benefit in the comprehension of the SARS-CoV-2 receptor recognition and its mechanism [13]. ACE-2 is a bioactive peptide that participates in the progression of cardiovascular diseases through the RAS system and effectually binds with SARS-CoV S protein [14,15]. The transmembrane spike glycoprotein has its domain (S1) exposed on the viral surface, and with it being enzymatically active, it mediates viral entry through the receptor-binding domain [16]. ACE-2 has been proven to be a specific functional receptor for SARS-CoV [14,17]. SARS-CoV-2 entry in cells without ACE-2 receptors, such as dipeptidyl peptidase 4 (DPP4), is impossible, whereas it easily enters ACE-2 receptor-expressing cells, which validates ACE-2 as the cell receptor for SARS-CoV-2 [18]. The fusion of the host cell and virus membrane causes the release of the viral RNA to the cytoplasm, resulting in infection. In SARS-CoV infection, ACE-2 is internalized along with the virus; however, its peptidase activity and catalytically active site are not distorted due to the binding process. Transmembrane proteinases (such as ADAM17 and TMPRSS2) and proteins (such as clathrin) are possibly involved in the binding and membrane fusion process [19,20,21]. A disintegrin and metallopeptidase domain 17 [ADAM17] leads to ectodomain shedding by cleaving to ACE-2, whereas transmembrane protease serine 2 [TMPRSS2] leads to viral uptake enhancement when it cleaves to ACE-2 [18,22]. Single-cell RNA-seq analysis and immuno-histochemical analysis application in various human cells disclose that the alveolar epithelial cells produce higher expression of ACE-2 than in oral and nasal mucosa epithelial cells, which blatantly points out the fact that the main target of SARS-CoV-2 is the lungs [23]. The heart may also be the first-hand target of SARS-CoV-2 because the cardiomyocytes, endothelial cells, and pericytes that are located in the heart express ACE-2 [15,24]. This has been well depicted, as per Figure 2 below.
SARS-CoV-2 downregulates the ACE-2 receptors. ACE-2 is significant in the degradation of angiotensin II, resulting in heptapeptide production, which is referred to as angiotensin 1–7. Therefore, an imbalance between ACE-2 and angiotensin II levels is noted. This imbalance is responsible for the cardiovascular complications in COVID-19 patients, including those with no prior CVD history and the aggravated condition in those with existing CVD. The anti-inflammatory and vasodilator effects on the cardiovascular system are observed when this heptapeptide binds to G protein-coupled receptor [25,26].
Oudit et al. demonstrated that SARS-CoV could affect the heart, resulting in ACE-2 protein level depletion [27]. Interestingly, autopsy reports from the hearts of SARS-CoV-2 infected patients showed notably depleted ACE-2 levels.

3.2. Cytokine Storm

Cytokine release syndrome (CRS) is an inflammatory response that is systemic, and it was first explained during the use of anti-T-cell antibody muromonab-CD3 (OKT3), a drug used after an organ transplantation has taken place [28]. CRS is meant to be a form of an inherent immune response, but it can become fatal when there are complications in various viral infections. Multiple organ failure and ARDS (acute respiratory distress syndrome) are some of the undesired effects of CRS. It is important to note as well that CRS was observed in both MERS and SARS epidemics as it is also being reported in SARS-CoV-2 [29]. CRS can either be mild, wherein the clinical manifestations include fatigue, rash, and arthralgia, or it can be severe, wherein the clinical manifestations will include organ failure, DIC (disseminated intravascular coagulopathy), vascular damage, and even shock [30].
CRS may cause cardiovascular complications by promoting cardiac toxicity, and the resultant cardiomyopathy resembles Takotsubo cardiomyopathy. Tachycardia, arrhythmia, and reduced ejection fraction may be experienced during this cardiac malfunction. The laboratory tests normally depict elevated C-reactive protein levels, distorted D-dimer values, and altered prothrombin time [29,31]. Laboratory investigations showed that lymphocytopenia in COVID-19 patients is linked with the severity of the symptoms manifested [32]. Contrary to expectation, the CRS fails to protect the body but destroys it instead through an exceeded immune response. Low cytokine and chemokine levels are initially produced after the COVID-19 infection in the respiratory epithelial cells and macrophages. Elevated proinflammatory cytokine levels (IL-1β, IL-6, and TNF) with reduced antiviral factors interferons (IFNs) levels are subsequently observed as the mechanism progresses. Inflammatory cells (neutrophils, monocytes) are enticed with high levels of chemokines and cytokines, causing lung tissue infiltration and, ultimately, lung injury. This macrophage and neutrophil intrusion into the lungs causes diffuse alveolar destruction, which culminates in ARDS. Studies have depicted that IL-6 levels correspond emphatically to disease severity, and drugs that inhibit IL-6 secretion should be considered in CRS treatment [33]. This has been well depicted, as per Figure 3 below.

3.3. Plaque Alteration

The elevated catecholamine levels due to systemic inflammation (as depicted in COVID-19 patients) may cause acute coronary syndrome by rapturing the plaque [34]. The C-reactive protein levels have a direct connection with the onset of myocardial infarction as a result of plaque rapture [35]. According to a study by Wang et al., elevated C-reactive protein in COVID-19 patients influences the severity of the clinical manifestation [36]. When the plaque ruptures, there is exposure to macrophages that are based under the endothelium. Microthrombi development begins due to the blood coming into contact with the expressed tissue factor [37]. Moreover, an acute response may be activated due to IL-6 over-production if the smooth muscle cells undergo inflammatory activation [38]. Figure 4 below shows this mechanism.

3.4. Prothrombotic State and Hypercoagulability

Microthrombi development occurs as a result of systemic inflammation, and it may lead to an impediment to the normal functioning of various organs. The hematopoietic system also undergoes restriction due to this inflammation, which is normally caused by infections [39].
Significantly elevated D-dimer levels (36%) and elevated incomplete thromboplastin time (6% of patients) were seen in a study involving 99 COVID-19-infected patients in Wuhan [40]. Moreover, a meta-analysis study showed that there was a strong correlation between D-dimer levels and the severity of COVID-19 in patients [41]. Another study was conducted to measure the coagulation parameters in 216 COVID-19-confirmed cases, and the results showed that 44 patients (20%) experienced prolonged aPTT time (activated partial thromboplastin clotting time). Lupus anticoagulant, which is linked with thrombotic tendency, was seen in 34 of these 44 patients [42]. Polyphosphates activate platelets and factor XII during systemic inflammation, and this ultimately leads to coagulation. Pathogen-induced mechanisms and elements of NETs (neutrophil extracellular traps) are also possibly involved in coagulation activation [38]. The initiation of endothelial inflammation throughout the systemic inflammatory response possibly enhances microthrombi formation as well [43].
Hypoxemia has procoagulant outcomes, and therefore it should also be contemplated as a possible cause of COVID-19 coagulation disorders. The diminished oxygen levels cause tissue factor transcription, resulting in vascular fibrin accumulation. Plasminogen activator inhibitor-1 is coactivated by hypoxia, and it acts by repressing fibrinolysis [44]. This has been well depicted, as per Figure 5 below. Additionally, Figure 6 shows a summary of the mechanisms involved in the development of cardiovascular complications in COVID-19.

4. Cardiovascular Complications of COVID-19

4.1. Myocardial Infarction

MI (myocardial infarction) primarily occurs due to an obstruction of blood flow to the heart, resulting in the death of the heart tissues, and it is also referred to as heart failure [45]. Systemic inflammatory response syndrome enhances the possibility of plaque rupture, as well as thrombus development, which ensues in either non-ST-elevation MI or ST-elevation MI [46]. Various mechanisms have been proposed to be responsible for AMI in COVID-19 patients. A proinflammatory state may cause further coronary plaque damage, as was observed in the influenza outbreak, resulting in type-1 AMI [47]. An imbalance between the lowered oxygen supply and the elevated oxygen demand in the heart may be accountable for type-2 AMI [48]. A study on SARS-hospitalized patients revealed that acute MI was responsible for two out of five deaths [49]. Another study proved that STEMI patients admitted during the COVID-19 period had a higher rate of experiencing adverse events, as well as severe clinical manifestations [50]. Notably, there have been conflicting outcomes from the various pieces of research that have been conducted on STEMI patients’ hospital admission during the COVID-19 pandemic. Those that reported a decline in AMI hospital admission during COVID-19 include [50,51], whereas there are some that reported an increase, such as [52,53]. Some of the likely explanations for this disparity include unequal global distribution admission rates, patient financial constraints, and economic instability in low GDP countries [54,55].
Early treatment and management delivery has been proposed to be an effective measure in preventing fatal outcomes in AMI patients with COVID-19 infection [56]. Table 1 below gives a summary of the available clinical evidence we found from our literature survey.

4.2. Myocarditis

Myocarditis is a condition where the heart muscles (myocardium) are inflamed, causing a myriad of clinical manifestations, such as chest pain, irregular heartbeats, and dyspnea. Notably, viral infections are mainly responsible for causing myocarditis in developed nations, whereas, in underdeveloped ones, both viruses and bacteria (diphtheriae) contribute to its pathogenesis [57]. The Dallas criteria (histopathological) and histological and immunological methods are currently used in diagnosis. The EMB diagnostic tool, which combines immunohistochemical and histopathological methods, is the best method in myocarditis diagnosis [58]. Interestingly, myocarditis was noted during the SARS-CoV and MERS-COV epidemics [59,60]. The first case reports of SARS-CoV-2 lacked myocarditis assessment, which probably had an effect on the understanding of this complication in COVID-19 patients. ECG modifications and anomalous changes in cardiac enzymes, which point to acute myocardial injury in COVID-19 patients, were observed in studies by Wang et al. and Doyen et al. [4,61]. The first myocarditis case was noted in a 63-year-old male who had no history of hypertension or any cardiac issue, and both his IL-6 and myocardial injury biomarkers (such as troponin I, NT-BNP) levels were elevated, as reported by Zeng et al. [62]. Subsequent cases linking COVID-19 to myocarditis in COVID-19 patients below the age of 40 include [63,64], and those above age 40 include [65,66]. Some autopsy reports also confirmed the presence of myocarditis in studies by Wichmann et al. and Rapkiewicz et al. [67,68]. However, another study revealed that the mortality rates in COVID-19 patients due to myocarditis were lower compared to other cardiac-related comorbidities [69]. The main mechanisms for myocarditis and myocardial injury are cardiomyocytes destruction due to viral entry, cytokine release syndrome, and hyperinflammation. Conflicting results have been observed in different pieces of research involving autopsy examinations of COVID-19 patients. Whether SARS-CoV-2 directly acts on cardiomyocytes or not is disputable; nevertheless, a direct effect on human stem cell-obtained cardiomyocytes has been proven [70]. This autopsy study analysis showed that the virus was not detected in the heart [71]. In contrast, a study involving 39 autopsy reports showed that there was myocardium viral presence in 24 cases [72]. Similarly, in another study, 1 out of the 23 cases showed viral RNA presence in the heart, along with lymphocytic myocarditis [73]. A report from England showed that eight children who had either been infected with COVID-19 previously had relatives who presented hyperinflammation, fever, and myocardial injury [74]. Other countries did report similar cases as well [75,76]. This prompted the naming of the condition as multisystem inflammatory syndrome in children (MIS-C).

4.3. Arrhythmia

Arrhythmia is among the early clinical manifestations of COVID-19, and it may suggest cardiac involvement once it is persistent. In a study by Liu et al. involving 137 patients, 7% of the patients had heart palpitations as a COVID-19 symptom [77]. A higher rate of arrhythmia (17%) was observed in another study Wang et al. involving positive COVID-19 cases [4]. Sinus tachycardia was the prevalent form of arrhythmia though the causal factor for this is yet to be known [78]. Atrial arrhythmias were recorded in 27.5% of COVID-19 ICU patients compared to zero in non-ICU patients [79]. Another study conducted by Hendren et al. on 700 COVID-19 patients showed that atrial fibrillation was linked with ICU admission [80]. Ventricular arrhythmias are also experienced in COVID-19 patients that are in a critical state [81]. The new development of tachyarrhythmia, followed by an increase in cardiac biomarkers in COVID-19 patients, may point to myocarditis [78,82].

4.4. Myocardial Interstitial Fibrosis

Elevated levels of myocardial fibrosis were observed in patients with COVID-19 as well as those who had recovered when magnetic resonance imaging of the myocardium was conducted. Furthermore, there seemed to be an association between these elevated levels and the severity of the COVID-19 disease [63,83]. Myocardial fibrosis may be influenced by various cardiac issues (such as heart failure and heart attack), but its classic development marker is enhanced fibroblast activation, which is due to higher measures of transforming growth factor β1 (TGF-β1) [84].
Interleukin-1 receptor-like 1 (also referred to as ST2 or IL1-RL1) is superficially located on the fibroblast cell, as well as on cardiomyocytes, and it binds to Interleukin-33 (IL-33). TGF-β1 activates IL1-RL1 into its dissolved form, which, on binding with IL-33, prevents the expected cardioprotective effect [85,86,87]. Myocardial fibrosis was also found to be a significant risk factor in determining the severity of cardiovascular complications in COVID-19 patients [88].
CMR detected diffuse fibrosis in a COVID-19-infected 45-year-old female with no myocarditis history, and palpitations were among the symptoms experienced 3 months post-COVID-19 infection [89]. This was confirmed by another report from a 49-year-old male who experienced dyspnea 6 weeks after the onset of COVID-19 clinical manifestation [90]. Furthermore, the autopsy results of 14 COVID-19 patients showed that six patients had focal cardiac fibrosis, although all had a history of MI [91]. However, some reports have been contradictory, such as in a case where edema was observed in two patients that possibly experienced myocarditis as a result of COVID-19, and the fibrosis was undetected [71].

4.5. Endothelial Cell Dysfunction and Vasculitis

Endothelial cells (ECs) are significant in the various pathologies of COVID-19 because they are involved in coagulation, immune response, and platelet functioning [92,93]. The existence of SARS-CoV-2 in the endothelial cells of various organs was seen in the postmortem examinations of COVID-19 patients [94]. Viral infections of the ECs can result in endothelial damage and the destruction of vascular structure, ultimately causing internal leakage. Hyperinflammation, together with hypercoagulability, was seen in COVID-19 patients whose ECs were dysfunctional [20,95].
Elevated ACE-2 expression was seen in the postmortems of infected patients, along with cell distension, disrupted endothelial structure, and the disorientation of cells from the membrane, which leads to platelet aggregation [96]. scRNA-Seq analysis has shown that the genes linked with cytokine production, leukocyte induction, and immunomodulation are seen in ECs, and notably, they have a greater expression in the lungs than in other organs, which may insinuate that ECs are associated with inflammation to an extent [97]. Structural interference with ECs causes the platelets to bind with the membrane, leading to thrombosis, which is also seen in COVID-19-infected patients [98]. Oxidative stress on the ECs may lead to COVID-19-induced cardiovascular complications. When this occurs along with inflammation, thrombus formation becomes the resultant effect [99].

4.6. Thromboembolism

Thrombotic occurrences were noted to be frequent, particularly in ICU-admitted COVID-19 patients. A 40% rate of thrombotic event occurrence was reported in this study, with venous thromboembolism being higher than arterial thromboembolism [100]. Other studies confirmed this reality of thrombotic events (TEs) in critically ill COVID-19 patients [101,102]. A postmortem examination of four COVID-19 patients showed the existence of sizeable emboli in the lungs, and other organs, such as the brain, had microthrombi [103]. Conspicuously, the existence of emboli has also been linked with increased severity of the condition and mortality as well [104,105,106,107]. TE aetiology is thought to be multifactorial, and this includes EC destruction, which causes collagen to be released, leading to platelet activation and, ultimately, thrombus formation. Additionally, hyperinflammation plays a role in mediating the onset of coagulopathy, as discussed earlier [95]. Other causes that may lead to a hypercoagulable state include hyperferritinemia, which is linked with macrophage activation syndrome and dispersed intravascular coagulation [39,108].

4.7. Dysautonomia

This is a medical state that is a result of a dysfunctional autonomic nervous system (ANS), resulting in the collapse of the parasympathetic, as well as sympathetic, components of the ANS, and its clinical manifestations include orthostatic intolerance, chest pain, and dizziness among others. It has been noted in COVID-19 patients, and it possibly occurs due to the progression of the infection or its prolongation (‘long’ COVID) [109,110,111]. Postural orthostatic hypotension (POTS), which is characterized by symptoms like fatigue, chest pain, and even orthostatic intolerance, may be experienced by some COVID-19 patients, and this is due to autonomic dysfunction [112,113,114]. Autoimmunity and hypovolemia, among others, could be the reason for POTS in COVID-19, although this is yet to be sufficiently verified [96,112].
Table 1. Summary of cardiovascular complications noted in COVID-19 patients.
Table 1. Summary of cardiovascular complications noted in COVID-19 patients.
Cardiovascular ComplicationAvailable Clinical EvidenceCause(s)
Myocardial infarctionTwo out of five SARS deaths are due to acute myocardial infarction (AMI) [49].Type 1 AMI- Inflammation
Type 2 AMI- Imbalance between the lowered oxygen supply and the elevated oxygen demand in the heart.
MyocarditisElevated IL-6 and myocardial injury biomarkers (such as troponin I and NT-BNP) levels were noted [64].Viral and bacterial infection.
Arrhythmia7–17% COVID-19 patients experienced arrhythmia [4,47].Myocardial inflammation.
Myocardial interstitial fibrosisElevated levels of myocardial fibrosis were detected, and it is used as a determinant of severity of cardiovascular complication in COVID-19 patients [63,83].Enhanced fibroblast activation due to higher measures of transforming growth factor β1 (TGF-β1) [84].
Endothelial cell dysfunction and VasculitisDisrupted endothelial structure was seen in the postmortems of infected COVID-19 patients [94].Oxidative stress on the endothelial cells.
ThromboembolismA study showed a 40% occurrence of thrombotic events [100].
Postmortems of four COVID-19 patients showed the existence of sizeable emboli in the lungs, brain, and other organs [103].
Platelet activation, hyperinflammation.
DysautonomiaNoted in COVID-19 patients [109,110,111].Prolongation (‘long’ COVID) infection.

5. Treatment

Table 2 below shows the various treatments that have been used in the treatment of COVID-19-induced cardiovascular complications with their outcomes, as seen from different case studies and reports.

5.1. Anticoagulant Therapy

Prophylactic anticoagulation is essential in the TE management of admitted COVID-19 patients. There has been a surge in both arterial thromboembolism- and venous thromboembolism (VTE)-hospitalized cases during the COVID-19 infection [102]. A study suggested that the VTE rate in hospitalized patients with COVID-19 may be higher than is contemplated [124]. However, the perfect thromboprophylaxis treatment is yet to be chosen. Figure 2 below has a summary of the currently available treatments.
Heparin was the earliest true anticoagulant to be discovered and it has been used in the treatment of conditions such as PE and DVT; however, some of its major drawbacks include bleeding, hyperkalaemia, and thrombocytopaenia [125].
A study involving 449 patients with acute COVID-19 revealed no changes in the 28-day mortality of 99 patients after low molecular weight heparin had been administered for a minimum of seven days, although changes were observed in the subgroups. The heparin subgroup experienced lesser mortality in comparison to the non-heparin group [126].
Enoxaparin was endorsed as the drug choice to be used in acute-phase VTE, whereas rivaroxaban and apixaban are to be chosen in postdischarge prophylaxis in those COVID-19 patients at a greater risk of developing thromboembolic events [127].

5.2. ACE Inhibitors and Angiotensin Receptor Blockers

SARS-CoV-2 binds to ACE-2 receptor prior to its entrance to the cell [20], and using the renin-angiotensin-aldosterone system (RAAS) inhibitors, which may raise ACE-2 expression, has raised doubts among researchers [128].
It is yet to be discovered if the greater expression of ACE-2 enhances predisposition to SARS-CoV-2 infection, but it was suggested that the use of RAAS inhibitors in COVID-19-stable patients should be continued [129].
The presence of divergent outcomes in both the preclinical and clinical studies that evaluated the effect of RAAS inhibition on ACE-2 expression has increased speculation [130]; however, this was clarified by a study that was carried out in the COVID-19 context. According to this study, it was made clear that various RAAS inhibitors have distinct effects on ACE-2 expression, hence the varying outcomes [131]. In a study conducted on mouse models, the decreased expression of ACE-2 showed a greater extent of lung damage caused by influenza [131]. Furthermore, in relation to COVID-19, a comprehensive study showed that the administration of RAS inhibitors in outpatients in no way enhanced the chances of COVID-19 diagnosis, its severity, or even the probability of hospitalization [132].
Angiotensin-receptor blockers and mineralocorticoid-receptor blockers enhance ACE-2 expression and activity both in preclinical and clinical studies [133]. However, ACE inhibitors only enhance expression but have no effect on activity, as seen in experimental models [130,131].

5.3. Immunosuppressive Therapy

The regular use of corticosteroids is not endorsed in SARS-CoV-2, as was the case in the SARS and MERS epidemics. This is because they may aggravate the already occurring lung injury [134]. Nonetheless, the injurious effects of the cytokine storm in COVID-19-infected patients have prompted the use of immunosuppression to control the hyperinflammatory response. Screening the infected patients and evaluating their laboratory values (such as platelet count and erythrocyte sedimentation rate) and also applying the use of hemophagocytic response scores, which are used in hyperinflammation diagnosis, may be beneficial in recognizing patients that need immunosuppression for an improved outcome [135,136].
IL-6 inhibitors, such as tocilizumab, may be useful as immunomodulators. In a case series involving 21 COVID-19 cases, tocilizumab ameliorated the symptoms, and it did prevent further worsening of the patients [137].
Furthermore, according to a study by Thakur et al., it was observed that the use of steroids among hospitalized patients proved to be effective as the number of COVID-19 deaths was reduced [138].

5.4. Mechanical Support

It was noted that some COVID-19 patients developed shock [4,139]. This prompted the need for mechanical circulatory support, and, when necessary, pulmonary support should be included [140]. Some of the techniques that should be used include left ventricular assist devices (LVADs) and venoarterial extracorporeal membrane oxygenation, which are very important, particularly in critical conditions of respiratory failure [141,142]. In the 2009 influenza epidemic, 13 myocarditis patients were assessed, and nine out of them, who were using an intra-aortic balloon pump (IABP), which is a mechanical circulatory support, survived, whereas the remaining four that were not on mechanical circulatory support succumbed [143]. A case has also been reported of a COVID-19 patient experiencing cardiogenic shock who recovered after being on IABP support [144]. The selection of patients who are to receive the various mechanical support systems should consider any other comorbidities being experienced by the patient in order to help prevent aggravating the patient’s condition.

5.5. Antiviral Drugs

Most of the drugs being used in the treatment of COVID-19 are similar to those used in the SARS and MERS epidemics. However, some new antiviral drugs, like chloroquine, have also been embedded in treatment to ensure enhanced, effective treatment. A blend of two or more antiviral drugs is not advocated for when considering toxicity, which may be incurred. There is still a lack of sufficient quality clinical evidence about this therapy, although a few studies have depicted a desirable outcome in COVID-19 patients having cardiovascular complications. An example of the antiviral drugs currently being used is briefly discussed below and shown in Figure 7.

Chloroquine and Hydroxychloroquine

These are basically meant for malarial treatment, and they are of the quinoline family. Additionally, they can be used in lupus erythematosus, as well as in rheumatoid arthritis treatment [145]. Chloroquine showed a positive outcome as a potential drug to be used in SARS, and recently, another study revealed that chloroquine could adequately impede SARS-CoV-2 by restraining ACE2 glycosylation by elevating the endosomal pH [146]. The Italian treatment guidelines approved the use of this combination, but the FDA, which had, earlier on, accepted this combination, revoked its emergency use after clinical trials had been conducted [147,148].

5.6. Antibiotics

Azithromycin

This is an antibiotic that inhibits inflammatory processes [149]. It was effective in both Zika virus and Ebola virus treatment [150]. It has been suggested that it should be used together with hydroxychloroquine, although the mechanism is not well established yet [151].
Chloroquine and azithromycin showed a positive outcome; however, their main adverse effect is QTc prolongation, which may be fatal, especially in COVID-19 patients. However, guidelines have been created with respect to handling Qt prolongation in COVID-19 patients [152].

6. Conclusions

The development of cardiovascular complications in COVID-19 patients worsened the pandemic situation. The lack of oxygen cylinders and the exacerbation of the symptoms, particularly in less developed nations, proved fatal. Therefore, this review was mainly meant to elucidate the proposed mechanisms in the pathogenesis of cardiovascular complications in COVID-19 patients, as well as the possible effective treatment modalities that have already been used and have had a notable positive therapeutic effect. Future studies should include larger cohort studies with prompt reporting of patients’ responses to the drugs. This will ultimately help better our comprehension of the complications and the effective treatments. Finally, there should be an increase in preclinical studies for better validifying the clinical results obtained.
We recommend that the knowledge and aptitude of the physicians be strengthened regularly through seminars, news, etc., on the latest advancements. Additionally, the judicious practice of medical ethics should be considered, especially in special conditions, such as comorbidities, terminally ill patients, etc.
A possible limitation of this study is that we did not use the traditional system of review, where all evidence is counted to be equal. On the other hand, our work favored research that has already had a high impact. Therefore, it is possible that we did not add some other references that are true but have less impact.

Author Contributions

I.O.: Conceptualization, Methodology, Writing—original draft preparation. M.V.: Data curation, Figure(s) preparation. M.A.: Visualization, Investigation. A.S.: Supervision, Review and Editing; P.K.: Validation. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank ISF College of Pharmacy, Moga for providing the necessary facilities required during this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Available online: https://www.who.int/news/item/29-06-2020-covidtimeline (accessed on 22 August 2022).
  2. Lu, R.; Zhao, X.; Li, J.; Niu, P.; Yang, B.; Wu, H.; Wang, W.; Song, H.; Huang, B.; Zhu, N.; et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding. Lancet 2020, 395, 565–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506, Erratum in Lancet 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical Characteristics of 138 Hospitalized Patients with 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA—J. Am. Med. Assoc. 2020, 323, 1061–1069, Erratum in JAMA 2021, 325, 1113. [Google Scholar] [CrossRef] [PubMed]
  5. Abdelrahman, Z.; Li, M.; Wang, X. Comparative Review of SARS-CoV-2, SARS-CoV, MERS-CoV, and Influenza A Respiratory Viruses. Front. Immunol. 2020, 11, 552909. [Google Scholar] [CrossRef]
  6. Novel Coronavirus Pneumonia Emergency Response Epidemiology Team. Consideration on the strategies during epidemic stage changing from emergency response to continuous prevention and control. Chin. J. Epidemiol. 2020, 41, 145–151. [Google Scholar] [CrossRef]
  7. Guo, T.; Fan, Y.; Chen, M.; Wu, X.; Zhang, L.; He, T.; Wang, H.; Wan, J.; Wang, X.; Lu, Z. Cardiovascular Implications of Fatal Outcomes of Patients with Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020, 5, 811–818, Erratum in JAMA Cardiol. 2020, 5, 848. [Google Scholar] [CrossRef] [Green Version]
  8. Wu, Z.; McGoogan, J.M. Characteristics of and Important Lessons from the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72 314 Cases from the Chinese Center for Disease Control and Prevention. JAMA 2020, 323, 1239–1242. [Google Scholar] [CrossRef]
  9. Bavishi, C.; Bonow, R.O.; Trivedi, V.; Abbott, J.D.; Messerli, F.H.; Bhatt, D.L. Special Article—Acute myocardial injury in patients hospitalized with COVID-19 infection: A review. Prog. Cardiovasc. Dis. 2020, 63, 682–689. [Google Scholar] [CrossRef]
  10. de Almeida-Pititto, B.; Dualib, P.M.; Zajdenverg, L.; Dantas, J.R.; de Souza, F.D.; Rodacki, M.; Bertoluci, M.C. Brazilian Diabetes Society Study Group (SBD) Severity and mortality of COVID-19 in patients with diabetes, hypertension and cardiovascular disease: A meta-analysis. Diabetol. Metab. Syndr. 2020, 12, 75. [Google Scholar] [CrossRef]
  11. Djaharuddin, I.; Munawwarah, S.; Nurulita, A.; Ilyas, M.; Tabri, N.A.; Lihawa, N. Comorbidities and mortality in COVID-19 patients. Gac. Sanit. 2020, 35, S530–S532. [Google Scholar] [CrossRef]
  12. Bae, S.; Kim, S.R.; Kim, M.-N.; Shim, W.J.; Park, S.-M. Impact of cardiovascular disease and risk factors on fatal outcomes in patients with COVID-19 according to age: A systematic review and meta-analysis. Heart 2021, 107, 373–380. [Google Scholar] [CrossRef] [PubMed]
  13. de Wit, E.; Van Doremalen, N.; Falzarano, D.; Munster, V.J. SARS and MERS: Recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 2016, 14, 523–534. [Google Scholar] [CrossRef] [PubMed]
  14. Sanchis-Gomar, F.; Lavie, C.J.; Perez-Quilis, C.; Henry, B.M.; Lippi, G. Angiotensin-Converting Enzyme 2 and Antihypertensives (Angiotensin Receptor Blockers and Angiotensin-Converting Enzyme Inhibitors) in Coronavirus Disease 2019. Mayo Clin. Proc. 2020, 95, 1222–1230. [Google Scholar] [CrossRef] [PubMed]
  15. Hamming, I.; Timens, W.; Bulthuis, M.L.C.; Lely, A.T.; Navis, G.J.; van Goor, H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. J. Pathol. 2004, 203, 631–637. [Google Scholar] [CrossRef]
  16. Ni, W.; Yang, X.; Yang, D.; Bao, J.; Li, R.; Xiao, Y.; Hou, C.; Wang, H.; Liu, J.; Yang, D.; et al. Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19. Crit. Care 2020, 24, 422. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, H.; Penninger, J.M.; Li, Y.; Zhong, N.; Slutsky, A.S. Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: Molecular mechanisms and potential therapeutic target. Intensive Care Med. 2020, 46, 586–590. [Google Scholar] [CrossRef] [Green Version]
  18. Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273, Erratum in Nature 2020, 588, E6. [Google Scholar] [CrossRef] [Green Version]
  19. Inoue, Y.; Tanaka, N.; Tanaka, Y.; Inoue, S.; Morita, K.; Zhuang, M.; Hattori, T.; Sugamura, K. Clathrin-Dependent Entry of Severe Acute Respiratory Syndrome Coronavirus into Target Cells Expressing ACE2 with the Cytoplasmic Tail Deleted. J. Virol. 2007, 81, 8722–8729. [Google Scholar] [CrossRef] [Green Version]
  20. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef]
  21. Li, F. Structure, Function, and Evolution of Coronavirus Spike Proteins. Annu. Rev. Virol. 2016, 3, 237–261. [Google Scholar] [CrossRef] [Green Version]
  22. Heurich, A.; Hofmann-Winkler, H.; Gierer, S.; Liepold, T.; Jahn, O.; Pöhlmann, S. TMPRSS2 and ADAM17 Cleave ACE2 Differentially and Only Proteolysis by TMPRSS2 Augments Entry Driven by the Severe Acute Respiratory Syndrome Coronavirus Spike Protein. J. Virol. 2014, 88, 1293–1307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ragia, G.; Manolopoulos, V.G. Inhibition of SARS-CoV-2 entry through the ACE2/TMPRSS2 pathway: A promising approach for uncovering early COVID-19 drug therapies. Eur. J. Clin. Pharmacol. 2020, 76, 1623–1630. [Google Scholar] [CrossRef] [PubMed]
  25. Der Sarkissian, S.; Grobe, J.; Yuan, L.; Narielwala, D.R.; Walter, G.A.; Katovich, M.J.; Raizada, M.K. Cardiac Overexpression of Angiotensin Converting Enzyme 2 Protects the Heart from Ischemia-Induced Pathophysiology. Hypertension 2008, 51, 712–718. [Google Scholar] [CrossRef] [Green Version]
  26. Zhong, J.; Basu, R.; Guo, D.; Chow, F.L.; Byrns, S.; Schuster, M.; Loibner, H.; Wang, X.-H.; Penninger, J.M.; Kassiri, Z.; et al. Angiotensin-Converting Enzyme 2 Suppresses Pathological Hypertrophy, Myocardial Fibrosis, and Cardiac Dysfunction. Circulation 2010, 122, 717–728. [Google Scholar] [CrossRef]
  27. Oudit, G.Y.; Kassiri, Z.; Jiang, C.; Liu, P.P.; Poutanen, S.; Penninger, J.; Butany, J. SARS-coronavirus modulation of myocardial ACE2 expression and inflammation in patients with SARS. Eur. J. Clin. Investig. 2009, 39, 618–625. [Google Scholar] [CrossRef]
  28. Chatenoud, L.; Ferran, C.; Legendre, C.; Thouard, I.; Merite, S.; Reuter, A.; Gevaert, Y.; Kreis, H.; Franchimont, P.; Bach, J.-F. In vivo cell activation following OKT3 administration: Systemic Cytokine Release and Modulation by Corticosteroids. Transplantation 1990, 49, 697–702. [Google Scholar] [CrossRef]
  29. Shimabukuro-Vornhagen, A.; Gödel, P.; Subklewe, M.; Stemmler, H.J.; Schlößer, H.A.; Schlaak, M.; Kochanek, M.; Böll, B.; von Bergwelt-Baildon, M.S. Cytokine release syndrome. J. Immunother. Cancer 2018, 6, 56. [Google Scholar] [CrossRef] [Green Version]
  30. STAT: Cytokine Release Syndrome. Clin. J. Oncol. Nurs. 2020, 24, 456. [CrossRef]
  31. Lee, D.W.; Gardner, R.A.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.C.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef] [Green Version]
  32. Cao, X. COVID-19: Immunopathology and its implications for therapy. Nat. Rev. Immunol. 2020, 20, 269–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Henderson, L.A.; Canna, S.W.; Schulert, G.S.; Volpi, S.; Lee, P.Y.; Kernan, K.F.; Caricchio, R.; Mahmud, S.; Hazen, M.M.; Halyabar, O.; et al. On the Alert for Cytokine Storm: Immunopathology in COVID-19. Arthritis Rheumatol. 2020, 72, 1059–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Alwaqfi, N.R.; Ibrahim, K.S. COVID-19: An update and cardiac involvement. J. Cardiothorac. Surg. 2020, 15, 239. [Google Scholar] [CrossRef]
  35. Ridker, P.M.; Cushman, M.; Stampfer, M.J.; Tracy, R.P.; Hennekens, C.H. Inflammation, Aspirin, and the Risk of Cardiovascular Disease in Apparently Healthy Men. N. Engl. J. Med. 1997, 336, 973–979. [Google Scholar] [CrossRef]
  36. Wang, L. C-reactive protein levels in the early stage of COVID-19. Med. Mal. Infect. 2020, 50, 332–334. [Google Scholar] [CrossRef]
  37. Esmon, C.T. Inflammation and thrombosis. J. Thromb. Haemost. 2003, 1, 1343–1348. [Google Scholar] [CrossRef] [PubMed]
  38. Velazquez-Salinas, L.; Verdugo-Rodriguez, A.; Rodriguez, L.L.; Borca, M.V. The Role of Interleukin 6 During Viral Infections. Front. Microbiol. 2019, 10, 1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Connors, J.M.; Levy, J.H. COVID-19 and its implications for thrombosis and anticoagulation. Blood 2020, 135, 2033–2040. [Google Scholar] [CrossRef]
  40. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
  41. Yu, H.-H.; Qin, C.; Chen, M.; Wang, W.; Tian, D.-S. D-dimer level is associated with the severity of COVID-19. Thromb. Res. 2020, 195, 219–225. [Google Scholar] [CrossRef]
  42. Bowles, L.; Platton, S.; Yartey, N.; Dave, M.; Lee, K.; Hart, D.P.; MacDonald, V.; Green, L.; Sivapalaratnam, S.; Pasi, K.J.; et al. Lupus Anticoagulant and Abnormal Coagulation Tests in Patients with COVID-19. N. Engl. J. Med. 2020, 383, 288–290. [Google Scholar] [CrossRef] [PubMed]
  43. Chang, J.C. Sepsis and septic shock: Endothelial molecular pathogenesis associated with vascular microthrombotic disease. Thromb. J. 2019, 17, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Price, L.C.; McCabe, C.; Garfield, B.; Wort, S.J. Thrombosis and COVID-19 pneumonia: The clot thickens! Eur. Respir. J. 2020, 56, 2001608. [Google Scholar] [CrossRef] [PubMed]
  45. Rathore, V.; Singh, N.; Mahat, R.K. Risk Factors of Acute Myocardial Infarction: A Review. Eurasian J. Med. Investig. 2018, 2, 1–7. [Google Scholar] [CrossRef]
  46. Warren-Gash, C.; Hayward, A.; Hemingway, H.; Denaxas, S.; Thomas, S.L.; Timmis, A.D.; Whitaker, H.; Smeeth, L. Influenza Infection and Risk of Acute Myocardial Infarction in England and Wales: A CALIBER Self-Controlled Case Series Study. J. Infect. Dis. 2012, 206, 1652–1659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Fosbøl, E.L.; Butt, J.H.; Østergaard, L.; Andersson, C.; Selmer, C.; Kragholm, K.; Schou, M.; Phelps, M.D.; Gislason, G.H.; Gerds, T.A.; et al. Association of Angiotensin-Converting Enzyme Inhibitor or Angiotensin Receptor Blocker Use with COVID-19 Diagnosis and Mortality. JAMA 2020, 324, 168–177. [Google Scholar] [CrossRef] [PubMed]
  48. Schiavone, M.; Gobbi, C.; Biondi-Zoccai, G.; D’Ascenzo, F.; Palazzuoli, A.; Gasperetti, A.; Mitacchione, G.; Viecca, M.; Galli, M.; Fedele, F.; et al. Acute Coronary Syndromes and COVID-19: Exploring the Uncertainties. J. Clin. Med. 2020, 9, 1683. [Google Scholar] [CrossRef]
  49. Peiris, J.; Chu, C.; Cheng, V.; Chan, K.; Hung, I.; Poon, L.; Law, K.; Tang, B.; Hon, T.; Chan, C.; et al. Clinical progression and viral load in a community outbreak of coronavirus-associated SARS pneumonia: A prospective study. Lancet 2003, 361, 1767–1772. [Google Scholar] [CrossRef] [Green Version]
  50. Fardman, A.; Zahger, D.; Orvin, K.; Oren, D.; Kofman, N.; Mohsen, J.; Tsafrir, O.; Asher, E.; Rubinshtein, R.; Jamal, J.; et al. Acute myocardial infarction in the COVID-19 era: Incidence, clinical characteristics and in-hospital outcomes—A multicenter registry. PLoS ONE 2021, 16, e0253524. [Google Scholar] [CrossRef]
  51. Tam, C.-C.F.; Cheung, K.-S.; Lam, S.; Wong, A.; Yung, A.; Sze, M.; Lam, Y.-M.; Chan, C.; Tsang, T.-C.; Tsui, M.; et al. Impact of Coronavirus Disease 2019 (COVID-19) Outbreak on ST-Segment–Elevation Myocardial Infarction Care in Hong Kong, China. Circ. Cardiovasc. Qual. Outcomes 2020, 13, e006631. [Google Scholar] [CrossRef]
  52. Harris, C.; Bisquera, A.; Lunt, A.; Peacock, J.L.; Greenough, A. Outcomes of the Neonatal Trial of High-Frequency Oscillation at 16 to 19 Years. N. Engl. J. Med. 2020, 383, 689–691. [Google Scholar] [CrossRef] [PubMed]
  53. Salinas, P.; Travieso, A.; Vergara-Uzcategui, C.; Tirado-Conte, G.; Macaya, F.; Mejía-Rentería, H.; Nombela-Franco, L.; Núñez-Gil, I.J.; Gonzalo, N.; Jiménez-Quevedo, P.; et al. Clinical Profile and 30-Day Mortality of Invasively Managed Patients with Suspected Acute Coronary Syndrome During the COVID-19 Outbreak. Int. Heart J. 2021, 62, 274–281. [Google Scholar] [CrossRef]
  54. Pessoa-Amorim, G.; Camm, C.F.; Gajendragadkar, P.; De Maria, G.L.; Arsac, C.; Laroche, C.; Zamorano, J.L.; Weidinger, F.; Achenbach, S.; Maggioni, A.P.; et al. Admission of patients with STEMI since the outbreak of the COVID-19 pandemic: A survey by the European Society of Cardiology. Eur. Heart J.—Qual. Care Clin. Outcomes 2020, 6, 210–216. [Google Scholar] [CrossRef]
  55. Smolderen, K. Health Care Insurance, Financial Concerns in Accessing Care, and Delays to Hospital Presentation in Acute Myocardial Infarction. JAMA 2010, 303, 1392–1400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Toscano, O.; Cosentino, N.; Campodonico, J.; Bartorelli, A.L.; Marenzi, G. Acute Myocardial Infarction During the COVID-19 Pandemic: An Update on Clinical Characteristics and Outcomes. Front. Cardiovasc. Med. 2021, 8, 648290. [Google Scholar] [CrossRef]
  57. Sagar, S.; Liu, P.P.; Cooper, L.T., Jr. Myocarditis. Rev. Del. Col. Am. Cardiol. 2012, 379, 738–747. [Google Scholar] [CrossRef] [Green Version]
  58. Pollack, A.; Kontorovich, A.; Fuster, V.; Dec, G.W. Viral myocarditis—Diagnosis, treatment options, and current controversies. Nat. Rev. Cardiol. 2015, 12, 670–680. [Google Scholar] [CrossRef] [PubMed]
  59. Chong, M.P.Y.; Chui, M.P.; Ling, M.A.E.; Franks, T.J.; Tai, M.D.Y.H.; Leo, M.Y.S.; Kaw, M.G.J.L.; Wansaicheong, M.G.; Chan, M.K.P.; Oon, M.L.L.E.; et al. Analysis of Deaths During the Severe Acute Respiratory Syndrome (SARS) Epidemic in Singapore: Challenges in Determining a SARS Diagnosis. Arch. Pathol. Lab. Med. 2004, 128, 195–204. [Google Scholar] [CrossRef]
  60. Alhogbani, T. Acute myocarditis associated with novel Middle East respiratory syndrome coronavirus. Ann. Saudi Med. 2016, 36, 78–80. [Google Scholar] [CrossRef] [Green Version]
  61. Doyen, D.; Moceri, P.; Ducreux, D.; Dellamonica, J. Myocarditis in a patient with COVID-19: A cause of raised troponin and ECG changes. Lancet 2020, 395, 1516. [Google Scholar] [CrossRef]
  62. Zeng, J.-H.; Liu, Y.-X.; Yuan, J.; Wang, F.-X.; Wu, W.-B.; Li, J.-X.; Wang, L.-F.; Gao, H.; Wang, Y.; Dong, C.-F.; et al. First case of COVID-19 complicated with fulminant myocarditis: A case report and insights. Infection 2020, 48, 773–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Clark, D.E.; Parikh, A.; Dendy, J.M.; Diamond, A.B.; George-Durrett, K.; Fish, F.A.; Slaughter, J.C.; Fitch, W.; Hughes, S.G.; Soslow, J.H. COVID-19 Myocardial Pathology Evaluation in Athletes With Cardiac Magnetic Resonance (COMPETE CMR). Circulation 2021, 143, 609–612. [Google Scholar] [CrossRef]
  64. Ludvigsson, J.F. Systematic review of COVID-19 in children shows milder cases and a better prognosis than adults. Acta Paediatr. 2020, 109, 1088–1095. [Google Scholar] [CrossRef]
  65. Inciardi, R.M.; Lupi, L.; Zaccone, G.; Italia, L.; Raffo, M.; Tomasoni, D.; Cani, D.S.; Cerini, M.; Farina, D.; Gavazzi, E.; et al. Cardiac Involvement in a Patient with Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020, 5, 819–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Coyle, J.; Igbinomwanhia, E.; Sanchez-Nadales, A.; Danciu, S.; Chu, C.; Shah, N. A Recovered Case of COVID-19 Myocarditis and ARDS Treated with Corticosteroids, Tocilizumab, and Experimental AT-001. JACC Case Rep. 2020, 2, 1331–1336. [Google Scholar] [CrossRef]
  67. Wichmann, D.; Sperhake, J.P.; Lütgehetmann, M.; Steurer, S.; Edler, C.; Heinemann, A.; Heinrich, F.; Mushumba, H.; Kniep, I.; Schröder, A.S.; et al. Autopsy Findings and Venous Thromboembolism in Patients with COVID-19. Ann. Intern. Med. 2020, 173, 268–277. [Google Scholar] [CrossRef] [PubMed]
  68. Rapkiewicz, A.V.; Mai, X.; Carsons, S.E.; Pittaluga, S.; Kleiner, D.E.; Berger, J.S.; Thomas, S.; Adler, N.M.; Charytan, D.M.; Gasmi, B.; et al. Megakaryocytes and platelet-fibrin thrombi characterize multi-organ thrombosis at autopsy in COVID-19: A case series. Eclinicalmedicine 2020, 24, 100434. [Google Scholar] [CrossRef] [PubMed]
  69. Halushka, M.K.; Heide, R.S.V. Myocarditis is rare in COVID-19 autopsies: Cardiovascular findings across 277 postmortem examinations. Cardiovasc. Pathol. 2020, 50, 107300. [Google Scholar] [CrossRef]
  70. Bailey, A.L.; Dmytrenko, O.; Greenberg, L.; Bredemeyer, A.L.; Ma, P.; Liu, J.; Penna, V.; Lai, L.; Winkler, E.S.; Sviben, S.; et al. SARS-CoV-2 Infects Human Engineered Heart Tissues and Models COVID-19 Myocarditis. JACC Basic Transl. Sci. 2021, 6, 331–345. [Google Scholar] [CrossRef]
  71. Wenzel, P.; Kopp, S.; Göbel, S.; Jansen, T.; Geyer, M.; Hahn, F.; Kreitner, K.-F.; Escher, F.; Schultheiss, H.-P.; Münzel, T. Evidence of SARS-CoV-2 mRNA in endomyocardial biopsies of patients with clinically suspected myocarditis tested negative for COVID-19 in nasopharyngeal swab. Cardiovasc. Res. 2020, 116, 1661–1663. [Google Scholar] [CrossRef]
  72. Lindner, D.; Fitzek, A.; Bräuninger, H.; Aleshcheva, G.; Edler, C.; Meissner, K.; Scherschel, K.; Kirchhof, P.; Escher, F.; Schultheiss, H.-P.; et al. Association of Cardiac Infection With SARS-CoV-2 in Confirmed COVID-19 Autopsy Cases. JAMA Cardiol. 2020, 5, 1281–1285. [Google Scholar] [CrossRef] [PubMed]
  73. Buja, L.M.; Wolf, D.A.; Zhao, B.; Akkanti, B.; McDonald, M.; Lelenwa, L.; Reilly, N.; Ottaviani, G.; Elghetany, M.T.; Trujillo, D.O.; et al. The emerging spectrum of cardiopulmonary pathology of the coronavirus disease 2019 (COVID-19): Report of 3 autopsies from Houston, Texas, and review of autopsy findings from other United States cities. Cardiovasc. Pathol. 2020, 48, 107233. [Google Scholar] [CrossRef] [PubMed]
  74. Riphagen, S.; Gomez, X.; Gonzalez-Martinez, C.; Wilkinson, N.; Theocharis, P. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet 2020, 395, 1607–1608. [Google Scholar] [CrossRef]
  75. Cabrero-Hernández, M.; García-Salido, A.; Leoz-Gordillo, I.; Alonso-Cadenas, J.A.; Gochi-Valdovinos, A.; Brabin, A.G.; Caro-Patón, G.D.L.; Nieto-Moro, M.; De-Azagra-Garde, A.M.; Serrano-González, A. Severe SARS-CoV-2 Infection in Children with Suspected Acute Abdomen: A Case Series from a Tertiary Hospital in Spain. Pediatr. Infect. Dis. J. 2020, 39, e195–e198. [Google Scholar] [CrossRef]
  76. Feldstein, L.R.; Rose, E.B.; Horwitz, S.M.; Collins, J.P.; Newhams, M.M.; Son, M.B.F.; Newburger, J.W.; Kleinman, L.C.; Heidemann, S.M.; Martin, A.A.; et al. Multisystem Inflammatory Syndrome in U.S. Children and Adolescents. N. Engl. J. Med. 2020, 383, 334–346. [Google Scholar] [CrossRef]
  77. Liu, K.; Fang, Y.-Y.; Deng, Y.; Liu, W.; Wang, M.-F.; Ma, J.-P.; Xiao, W.; Wang, Y.-N.; Zhong, M.-H.; Li, C.-H.; et al. Clinical characteristics of novel coronavirus cases in tertiary hospitals in Hubei Province. Chin. Med. J. 2020, 133, 1025–1031. [Google Scholar] [CrossRef] [PubMed]
  78. Driggin, E.; Madhavan, M.V.; Bikdeli, B.; Chuich, T.; Laracy, J.; Biondi-Zoccai, G.; Brown, T.S.; Der Nigoghossian, C.; Zidar, D.A.; Haythe, J.; et al. Cardiovascular Considerations for Patients, Health Care Workers, and Health Systems During the COVID-19 Pandemic. J. Am. Coll. Cardiol. 2020, 75, 2352–2371. [Google Scholar] [CrossRef]
  79. Colon, C.M.; Barrios, J.G.; Chiles, J.W.; McElwee, S.K.; Russell, D.W.; Maddox, W.R.; Kay, G.N. Atrial Arrhythmias in COVID-19 Patients. JACC Clin. Electrophysiol. 2020, 6, 1189–1190. [Google Scholar] [CrossRef]
  80. Hendren, N.S.; Drazner, M.H.; Bozkurt, B.; Cooper, L.T., Jr. Description and Proposed Management of the Acute COVID-19 Cardiovascular Syndrome. Circulation 2020, 141, 1903–1914. [Google Scholar] [CrossRef] [Green Version]
  81. Chen, Q.; Xu, L.; Dai, Y.; Ling, Y.; Mao, J.; Qian, J.; Zhu, W.; Di, W.; Ge, J. Cardiovascular manifestations in severe and critical patients with COVID-19. Clin. Cardiol. 2020, 43, 796–802. [Google Scholar] [CrossRef]
  82. Chen, C.; Zhou, Y.; Wang, D.W. SARS-CoV-2: A potential novel etiology of fulminant myocarditis. Herz 2020, 45, 230–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Puntmann, V.O.; Carerj, M.L.; Wieters, I.; Fahim, M.; Arendt, C.; Hoffmann, J.; Shchendrygina, A.; Escher, F.; Vasa-Nicotera, M.; Zeiher, A.M.; et al. Outcomes of Cardiovascular Magnetic Resonance Imaging in Patients Recently Recovered from Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020, 5, 1265. [Google Scholar] [CrossRef] [PubMed]
  84. Khalil, H.; Kanisicak, O.; Prasad, V.; Correll, R.N.; Fu, X.; Schips, T.; Vagnozzi, R.J.; Liu, R.; Huynh, T.; Lee, S.-J.; et al. Fibroblast-specific TGF-β–Smad2/3 signaling underlies cardiac fibrosis. J. Clin. Investig. 2017, 127, 3770–3783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kotsiou, O.S.; Gourgoulianis, K.I.; Zarogiannis, S.G. IL-33/ST2 Axis in Organ Fibrosis. Front. Immunol. 2018, 9, 2432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Weinberg, E.; Shimpo, M.; De Keulenaer, G.; MacGillivray, C.; Tominaga, S.-I.; Solomon, S.D.; Rouleau, J.-L.; Lee, R.T. Expression and Regulation of ST2, an Interleukin-1 Receptor Family Member, in Cardiomyocytes and Myocardial Infarction. Circulation 2002, 106, 2961–2966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Matilla, L.; Arrieta, V.; Jover, E.; Garcia-Peña, A.; Martinez-Martinez, E.; Sadaba, R.; Alvarez, V.; Navarro, A.; Fernandez-Celis, A.; Gainza, A.; et al. Soluble St2 Induces Cardiac Fibroblast Activation and Collagen Synthesis via Neuropilin-1. Cells 2020, 9, 1667. [Google Scholar] [CrossRef]
  88. Mustroph, J.; Hupf, J.; Baier, M.J.; Evert, K.; Brochhausen, C.; Broeker, K.; Meindl, C.; Seither, B.; Jungbauer, C.; Evert, M.; et al. Cardiac Fibrosis Is a Risk Factor for Severe COVID-19. Front. Immunol. 2021, 12, 740260. [Google Scholar] [CrossRef]
  89. Jagia, P.; Ojha, V.; Naik, N.; Sharma, S. Myocardial fibrosis detected by cardiovascular magnetic resonance in absence of myocardial oedema in a patient recovered from COVID-19. BMJ Case Rep. 2020, 13, e240193. [Google Scholar] [CrossRef]
  90. Spano, G.; Fischer, K.; Maillat, C.; Vicario, G.; Huber, A.T.; Gräni, C. Delayed isolated peri-myocarditis in a COVID-19 patient with respiratory symptoms but without lung involvement. Int. J. Cardiovasc. Imaging 2020, 36, 2279–2280. [Google Scholar] [CrossRef]
  91. Grosse, C.; Grosse, A.; Salzer, H.J.; Dünser, M.W.; Motz, R.; Langer, R. Analysis of cardiopulmonary findings in COVID-19 fatalities: High incidence of pulmonary artery thrombi and acute suppurative bronchopneumonia. Cardiovasc. Pathol. 2020, 49, 107263. [Google Scholar] [CrossRef]
  92. Siddiqi, H.K.; Libby, P.; Ridker, P.M. COVID-19—A vascular disease. Trends Cardiovasc. Med. 2020, 31, 1–5. [Google Scholar] [CrossRef] [PubMed]
  93. Libby, P.; Lüscher, T. COVID-19 is, in the end, an endothelial disease. Eur. Heart J. 2020, 41, 3038–3044. [Google Scholar] [CrossRef] [PubMed]
  94. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef] [PubMed]
  95. Teuwen, L.-A.; Geldhof, V.; Pasut, A.; Carmeliet, P. COVID-19: The vasculature unleashed. Nat. Rev. Immunol. 2020, 20, 389–391, Erratum in Nat. Rev. Immunol. 2020, 20, 448. [Google Scholar] [CrossRef] [PubMed]
  96. Farshidfar, F.; Koleini, N.; Ardehali, H. Cardiovascular complications of COVID-19. J. Clin. Investig. 2021, 6, 1504–1507. [Google Scholar] [CrossRef]
  97. Goveia, J.; Rohlenova, K.; Taverna, F.; Treps, L.; Conradi, L.-C.; Pircher, A.; Geldhof, V.; de Rooij, L.P.; Kalucka, J.; Sokol, L.; et al. An Integrated Gene Expression Landscape Profiling Approach to Identify Lung Tumor Endothelial Cell Heterogeneity and Angiogenic Candidates. Cancer Cell 2020, 37, 21–36.e13. [Google Scholar] [CrossRef] [PubMed]
  98. Panigada, M.; Bottino, N.; Tagliabue, P.; Grasselli, G.; Novembrino, C.; Chantarangkul, V.; Pesenti, A.; Peyvandi, F.; Tripodi, A. Hypercoagulability of COVID-19 patients in intensive care unit: A report of thromboelastography findings and other parameters of hemostasis. J. Thromb. Haemost. 2020, 18, 1738–1742. [Google Scholar] [CrossRef]
  99. Chang, R.; Mamun, A.; Dominic, A.; Le, N.-T. SARS-CoV-2 Mediated Endothelial Dysfunction: The Potential Role of Chronic Oxidative Stress. Front. Physiol. 2021, 11, 605908. [Google Scholar] [CrossRef]
  100. Fraissé, M.; Logre, E.; Pajot, O.; Mentec, H.; Plantefève, G.; Contou, D. Thrombotic and hemorrhagic events in critically ill COVID-19 patients: A French monocenter retrospective study. Crit. Care 2020, 24, 275. [Google Scholar] [CrossRef]
  101. Cui, S.; Chen, S.; Li, X.; Liu, S.; Wang, F. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. J. Thromb. Haemost. 2020, 18, 1421–1424. [Google Scholar] [CrossRef] [Green Version]
  102. Klok, F.A.; Kruip, M.J.H.A.; van der Meer, N.J.M.; Arbous, M.S.; Gommers, D.A.M.P.J.; Kant, K.M.; Kaptein, F.H.J.; van Paassen, J.; Stals, M.A.M.; Huisman, M.V.; et al. Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb. Res. 2020, 191, 145–147. [Google Scholar] [CrossRef] [PubMed]
  103. Bryce, C.; Grimes, Z.; Pujadas, E.; Ahuja, S.; Beasley, M.B.; Albrecht, R.; Hernandez, T.; Stock, A.; Zhao, Z.; AlRasheed, M.R.; et al. Pathophysiology of SARS-CoV-2: The Mount Sinai COVID-19 autopsy experience. Mod. Pathol. 2021, 34, 1456–1467. [Google Scholar] [CrossRef]
  104. Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef] [PubMed]
  105. Middeldorp, S.; Coppens, M.; van Haaps, T.F.; Foppen, M.; Vlaar, A.P.; Müller, M.C.; Bouman, C.C.; Beenen, L.F.; Kootte, R.S.; Heijmans, J.; et al. Incidence of venous thromboembolism in hospitalized patients with COVID-19. J. Thromb. Haemost. 2020, 18, 1995–2002. [Google Scholar] [CrossRef] [PubMed]
  106. Helms, J.; Tacquard, C.; Severac, F.; Leonard-Lorant, I.; Ohana, M.; Delabranche, X.; Merdji, H.; Clere-Jehl, R.; Schenck, M.; Gandet, F.F.; et al. High risk of thrombosis in patients with severe SARS-CoV-2 infection: A multicenter prospective cohort study. Intensive Care Med. 2020, 46, 1089–1098. [Google Scholar] [CrossRef] [PubMed]
  107. Shi, S.; Qin, M.; Shen, B.; Cai, Y.; Liu, T.; Yang, F.; Gong, W.; Liu, X.; Liang, J.; Zhao, Q.; et al. Association of Cardiac Injury With Mortality in Hospitalized Patients With COVID-19 in Wuhan, China. JAMA Cardiol. 2020, 5, 802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Lodigiani, C.; Iapichino, G.; Carenzo, L.; Cecconi, M.; Ferrazzi, P.; Sebastian, T.; Kucher, N.; Studt, J.-D.; Sacco, C.; Bertuzzi, A.; et al. Venous and arterial thromboembolic complications in COVID-19 patients admitted to an academic hospital in Milan, Italy. Thromb. Res. 2020, 191, 9–14. [Google Scholar] [CrossRef]
  109. Miglis, M.G.; Prieto, T.; Shaik, R.; Muppidi, S.; Sinn, D.-I.; Jaradeh, S. A case report of postural tachycardia syndrome after COVID-19. Clin. Auton. Res. 2020, 30, 449–451. [Google Scholar] [CrossRef]
  110. Kanjwal, K.; Jamal, S.; Kichloo, A.; Grubb, B. New-onset Postural Orthostatic Tachycardia Syndrome Following Coronavirus Disease 2019 Infection. J. Innov. Card. Rhythm. Manag. 2020, 11, 4302–4304. [Google Scholar] [CrossRef]
  111. Umapathi, T.; Poh, M.Q.W.; Fan, B.E.; Li, K.F.C.; George, J.; Tan, J.Y.L. Acute hyperhidrosis and postural tachycardia in a COVID-19 patient. Clin. Auton. Res. 2020, 30, 571–573. [Google Scholar] [CrossRef]
  112. Goldstein, D.S. The possible association between COVID-19 and postural tachycardia syndrome. Heart Rhythm. 2020, 18, 508–509. [Google Scholar] [CrossRef] [PubMed]
  113. Blitshteyn, S.; Whitelaw, S. Postural orthostatic tachycardia syndrome (POTS) and other autonomic disorders after COVID-19 infection: A case series of 20 patients. Immunol. Res. 2021, 69, 205–211. [Google Scholar] [CrossRef] [PubMed]
  114. Johansson, M.; Ståhlberg, M.; Runold, M.; Nygren-Bonnier, M.; Nilsson, J.; Olshansky, B.; Bruchfeld, J.; Fedorowski, A. Long-Haul Post–COVID-19 Symptoms Presenting as a Variant of Postural Orthostatic Tachycardia Syndrome. JACC Case Rep. 2021, 3, 573–580. [Google Scholar] [CrossRef]
  115. Bainey, K.; Bates, E.R.; Armstrong, P.W. ST-Segment-Elevation Myocardial Infarction Care and COVID-19: The Value Proposition of Fibrinolytic Therapy and the Pharmacoinvasive Strategy. Circ. Cardiovasc. Qual. Outcomes 2020, 13, e006834. [Google Scholar] [CrossRef] [PubMed]
  116. Othman, F.; Abid, A.R.; Alibrahim, M.; Abdulkarim, S.; Abdelaty, M.A.; Aboukamar, M.; Arafah, S. Non-ST Segment Elevation Myocardial Infarction in a Patient with COVID-19. Heart Views 2020, 21, 215–219. [Google Scholar] [CrossRef]
  117. Sala, S.; Peretto, G.; Gramegna, M.; Palmisano, A.; Villatore, A.; Vignale, D.; De Cobelli, F.; Tresoldi, M.; Cappelletti, A.M.; Basso, C.; et al. Acute myocarditis presenting as a reverse Tako-Tsubo syndrome in a patient with SARS-CoV-2 respiratory infection. Eur. Heart J. 2020, 41, 1861–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Ling, Y.; Xu, S.-B.; Lin, Y.-X.; Tian, D.; Zhu, Z.-Q.; Dai, F.-H.; Wu, F.; Song, Z.-G.; Huang, W.; Chen, J.; et al. Persistence and clearance of viral RNA in 2019 novel coronavirus disease rehabilitation patients. Chin. Med. J. 2020, 133, 1039–1043. [Google Scholar] [CrossRef]
  119. Lampropoulos, C.E.; Mavrogeni, S.; Dervas, A.; Manios, E.; Chatzidou, S.; Kontogiannis, C.; Spiliopoulou, S.; Melliou, S.; Kalogianni, E.; Papaioannou, I. Myocardial fibrosis after COVID-19 infection and severe sinus arrest episodes in an asymptomatic patient with mild sleep apnea syndrome: A case report and review of the literature. Respir. Med. Case Rep. 2021, 32, 101366. [Google Scholar] [CrossRef]
  120. Jin, Y.; Ji, W.; Yang, H.; Chen, S.; Zhang, W.; Duan, G. Endothelial activation and dysfunction in COVID-19: From basic mechanisms to potential therapeutic approaches. Signal Transduct. Target. Ther. 2020, 5, 293. [Google Scholar] [CrossRef]
  121. Hassan, W.; Ramadan, H.K.-A. COVID-19 as a novel etiology of portal vein thrombosis: Change in the current management concepts. Infect. Dis. 2020, 53, 148–150. [Google Scholar] [CrossRef]
  122. Desai, A.D.; Boursiquot, B.C.; Melki, L.; Wan, E.Y. Management of Arrhythmias Associated with COVID-19. Curr. Cardiol. Rep. 2020, 23, 2. [Google Scholar] [CrossRef] [PubMed]
  123. Carmona-Torre, F.; Mínguez-Olaondo, A.; López-Bravo, A.; Tijero, B.; Grozeva, V.; Walcker, M.; Azkune-Galparsoro, H.; de Munain, A.L.; Alcaide, A.B.; Quiroga, J.; et al. Dysautonomia in COVID-19 Patients: A Narrative Review on Clinical Course, Diagnostic and Therapeutic Strategies. Front. Neurol. 2022, 13, 886609. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, Z.-R.; Zhou, Y.; Liu, J.; Peng, H.-W.; Zhou, J.; Zhong, H.-L.; Liu, L.-L.; Lai, M.-F.; Wei, X.-H.; Wen, J.-H. Pharmacotherapics Advice in Guidelines for COVID-19. Front. Pharmacol. 2020, 11, 950. [Google Scholar] [CrossRef] [PubMed]
  125. Ahmed, I.; Majeed, A.; Powell, R. Heparin induced thrombocytopenia: Diagnosis and management update. Postgrad. Med. J. 2007, 83, 575–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Tang, N.; Bai, H.; Chen, X.; Gong, J.; Li, D.; Sun, Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J. Thromb. Haemost. 2020, 18, 1094–1099. [Google Scholar] [CrossRef] [Green Version]
  127. Chandra, A.; Chakraborty, U.; Ghosh, S.; Dasgupta, S. Anticoagulation in COVID-19: Current concepts and controversies. Postgrad. Med. J. 2021, 98, 395–402. [Google Scholar] [CrossRef]
  128. Esler, M.; Esler, D. Can angiotensin receptor-blocking drugs perhaps be harmful in the COVID-19 pandemic? J. Hypertens. 2020, 38, 781–782. [Google Scholar] [CrossRef]
  129. Danser, A.H.J.; Epstein, M.; Batlle, D. Renin-Angiotensin System Blockers and the COVID-19 Pandemic: At Present There Is No Evidence to Abandon Renin-Angiotensin System Blockers. Hypertension. 2020, 75, 1382–1385. [Google Scholar] [CrossRef] [Green Version]
  130. Ferrario, C.M.; Jessup, J.; Chappell, M.C.; Averill, D.B.; Brosnihan, K.B.; Tallant, E.A.; Diz, D.I.; Gallagher, P.E. Effect of Angiotensin-Converting Enzyme Inhibition and Angiotensin II Receptor Blockers on Cardiac Angiotensin-Converting Enzyme 2. Circulation 2005, 111, 2605–2610. [Google Scholar] [CrossRef] [Green Version]
  131. Zheng, Y.-Y.; Ma, Y.-T.; Zhang, J.-Y.; Xie, X. Reply to: ‘Interaction between RAAS inhibitors and ACE2 in the context of COVID-19′. Nat. Rev. Cardiol. 2020, 17, 313–314. [Google Scholar] [CrossRef] [Green Version]
  132. de Abajo, F.J. Renin–angiotensin system inhibitors and COVID-19: Overwhelming evidence against an association. Lancet Digit. Health 2020, 3, e70–e71. [Google Scholar] [CrossRef] [PubMed]
  133. Keidar, S.; Gamliel-Lazarovich, A.; Kaplan, M.; Pavlotzky, E.; Hamoud, S.; Hayek, T.; Karry, R.; Abassi, Z. Mineralocorticoid Receptor Blocker Increases Angiotensin-Converting Enzyme 2 Activity in Congestive Heart Failure Patients. Circ. Res. 2005, 97, 946–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Russell, C.D.; Millar, J.E.; Baillie, J.K. Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury. Lancet 2020, 395, 473–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J.; HLH Across Speciality Collaboration UK. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef] [PubMed]
  136. Fardet, L.; Galicier, L.; Lambotte, O.; Marzac, C.; Aumont, C.; Chahwan, D.; Coppo, P.; Hejblum, G. Development and Validation of the HScore, a Score for the Diagnosis of Reactive Hemophagocytic Syndrome. Arthritis Rheumatol. 2014, 66, 2613–2620. [Google Scholar] [CrossRef] [PubMed]
  137. Xu, X.; Han, M.; Li, T.; Sun, W.; Wang, D.; Fu, B.; Zhou, Y.; Zheng, X.; Yang, Y.; Li, X.; et al. Effective Treatment of Severe COVID-19 Patients with Tocilizumab. Proc. Natl. Acad. Sci. USA 2020, 117, 10970–10975. [Google Scholar] [CrossRef] [PubMed]
  138. Thakur, M.; Datusalia, A.K.; Kumar, A. Use of steroids in COVID-19 patients: A meta-analysis. Eur. J. Pharmacol. 2021, 914, 174579. [Google Scholar] [CrossRef]
  139. Clerkin, K.J.; Fried, J.A.; Raikhelkar, J.; Sayer, G.; Griffin, J.M.; Masoumi, A.; Jain, S.S.; Burkhoff, D.; Kumaraiah, D.; Rabbani, L.R.; et al. COVID-19 and Cardiovascular Disease. Circulation 2020, 141, 1648–1655. [Google Scholar] [CrossRef] [Green Version]
  140. Miller, P.E.; Solomon, M.A.; McAreavey, D. Advanced Percutaneous Mechanical Circulatory Support Devices for Cardiogenic Shock. Crit. Care Med. 2017, 45, 1922–1929. [Google Scholar] [CrossRef]
  141. Rajagopal, K.; Keller, S.P.; Akkanti, B.; Bime, C.; Loyalka, P.; Cheema, F.H.; Zwischenberger, J.B.; El Banayosy, A.; Pappalardo, F.; Slaughter, M.S.; et al. Advanced Pulmonary and Cardiac Support of COVID-19 Patients. Circ. Heart Fail. 2020, 13, e007175. [Google Scholar] [CrossRef]
  142. Osoro, I.; Sharma, A.; Amir, M.; Vohra, M.; Kumar, R.; Kumar, H.; Zargar, A.; Bangar, H. Prevention and management of anthracycline induced cardiotoxicity: A review. Health Sci. Rev. 2022, 5, 100070. [Google Scholar] [CrossRef]
  143. Ukimura, A.; Ooi, Y.; Kanzaki, Y.; Inomata, T.; Izumi, T. A national survey on myocarditis associated with influenza H1N1pdm2009 in the pandemic and postpandemic season in Japan. J. Infect. Chemother. 2013, 19, 426–431. [Google Scholar] [CrossRef] [PubMed]
  144. Fried, J.A.; Ramasubbu, K.; Bhatt, R.; Topkara, V.K.; Clerkin, K.J.; Horn, E.; Rabbani, L.; Brodie, D.; Jain, S.S.; Kirtane, A.J.; et al. The Variety of Cardiovascular Presentations of COVID-19. Circulation 2020, 141, 1930–1936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Schrezenmeier, E.; Dörner, T. Mechanisms of action of hydroxychloroquine and chloroquine: Implications for rheumatology. Nat. Rev. Rheumatol. 2020, 16, 155–166. [Google Scholar] [CrossRef] [PubMed]
  146. Wang, M.; Cao, R.; Zhang, L.; Yang, X.; Liu, J.; Xu, M.; Shi, Z.; Hu, Z.; Zhong, W.; Xiao, G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020, 30, 269–271. [Google Scholar] [CrossRef]
  147. Lombardy Section Italian Society Infectious And Tropical Diseases. Vademecum for the treatment of people with COVID-19. Edition 2.0, 13 March 2020. Infez Med. 2020, 28, 143–152. [Google Scholar]
  148. U.S Food and Drug Administration (FDA). Available online: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-revokes-emergency-use-authorization-chloroquine-and (accessed on 15 September 2022).
  149. Simoens, S.; Laekeman, G.; Decramer, M. Preventing COPD exacerbations with macrolides: A review and budget impact analysis. Respir. Med. 2013, 107, 637–648. [Google Scholar] [CrossRef] [Green Version]
  150. Retallack, H.; Di Lullo, E.; Arias, C.; Knopp, K.A.; Laurie, M.T.; Sandoval-Espinosa, C.; Mancia Leon, W.R.; Krencik, R.; Ullian, E.M.; Spatazza, J.; et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc. Natl. Acad. Sci. USA 2016, 113, 14408–14413. [Google Scholar] [CrossRef] [Green Version]
  151. Gautret, P.; Lagier, J.C.; Parola, P.; Hoang, V.T.; Meddeb, L.; Mailhe, M.; Doudier, B.; Courjon, J.; Giordanengo, V.; Vieira, V.E.; et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: Results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents 2020, 56, 105949. [Google Scholar] [CrossRef]
  152. Lakkireddy, D.R.; Chung, M.K.; Gopinathannair, R.; Patton, K.K.; Gluckman, T.J.; Turagam, M.; Cheung, J.W.; Patel, P.; Sotomonte, J.; Lampert, R.; et al. Guidance for cardiac electrophysiology during the COVID-19 pandemic from the Heart Rhythm Society COVID-19 Task Force; Electrophysiology Section of the American College of Cardiology; and the Electrocardiography and Arrhythmias Committee of the Council on Clinical Cardiology, American Heart Association. Heart Rhythm. 2020, 17, e233–e241. [Google Scholar] [CrossRef]
Figure 1. Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA-P).
Figure 1. Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols (PRISMA-P).
Covid 03 00026 g001
Figure 2. Diagrammatic illustration ofACE-2 inhibitors downregulation mechanism.
Figure 2. Diagrammatic illustration ofACE-2 inhibitors downregulation mechanism.
Covid 03 00026 g002
Figure 3. Diagrammatic illustration of cytokine storm mechanism.
Figure 3. Diagrammatic illustration of cytokine storm mechanism.
Covid 03 00026 g003
Figure 4. Diagrammatic illustration of plaque alteration mechanism.
Figure 4. Diagrammatic illustration of plaque alteration mechanism.
Covid 03 00026 g004
Figure 5. Diagrammatic illustration of prothrombotic state and hypercoagulability mechanism.
Figure 5. Diagrammatic illustration of prothrombotic state and hypercoagulability mechanism.
Covid 03 00026 g005
Figure 6. The possible mechanisms involved in the development of COVID-19 cardiovascular complications.
Figure 6. The possible mechanisms involved in the development of COVID-19 cardiovascular complications.
Covid 03 00026 g006
Figure 7. Currently available treatments.
Figure 7. Currently available treatments.
Covid 03 00026 g007
Table 2. Treatment consideration table.
Table 2. Treatment consideration table.
DiseaseTreatmentOutcomes
Myocardial infarctionSTEMI—(fibrinolytic therapy along with pharmacoinvasive strategy) [115]
NSTEMI—aspirin, clopidogrel, enoxaparin, noradrenaline [116].
Fibrinolytic therapy terminates MI
Patient showed good therapeutic response within 2 days and was finally discharged after 22 days though some complications arose during the treatment.
MyocarditisLopinavir/ritonavir/hydroxychloroquine [117]
IV prednisolone, and dexamethasone [118].
ECG normalized, LVEF returned to 65%, patient discharged after 13 days.
Prolonged duration before detection of viral RNA.
Myocardial interstitial fibrosisContinuous positive airway pressure (CPAP) with a possibility of pacemaker implantation [119].N/A
ECs dysfunction and vasculitisN-Acetyl-L-cysteine, Human recombinant IL-37 (antioxidant treatment)
targeting IL-6/IL-6R [120].
Lowered ROS formation
Lowered incidents of pathogenesis of COVID-19.
Thrombotic eventsAnticoagulation therapy + enoxaparin [121]
Anti-coagulant therapy+ heparin [121].
Patient was relieved of the abdominal pain and other symptoms
There was improvement in the health of all the patients except one that died.
ArrhythmiasAntiarrhythmic agents (amiodarone) + procainamide or lidocaine
If the patient is critical, defibrillation should be considered [122].
Improved patient health.
DysautonomiaHeart rate inhibitors (ivabradine)
Sympatholytic drugs clonidine and methyldopa)
Volume expanders (fludrocortisone and intravenous saline) [123].
Quick betterment of the symptoms and lowered heart rate.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Osoro, I.; Vohra, M.; Amir, M.; Kumar, P.; Sharma, A. A Scoping Review on COVID-19-Induced Cardiovascular Complications. COVID 2023, 3, 348-369. https://doi.org/10.3390/covid3030026

AMA Style

Osoro I, Vohra M, Amir M, Kumar P, Sharma A. A Scoping Review on COVID-19-Induced Cardiovascular Complications. COVID. 2023; 3(3):348-369. https://doi.org/10.3390/covid3030026

Chicago/Turabian Style

Osoro, Ian, Manisha Vohra, Mohammad Amir, Puneet Kumar, and Amit Sharma. 2023. "A Scoping Review on COVID-19-Induced Cardiovascular Complications" COVID 3, no. 3: 348-369. https://doi.org/10.3390/covid3030026

Article Metrics

Back to TopTop