Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us Membership Collaborators Partners
OPEN ACCESS

Potential of Phytomedicine in Benefiting Both Long COVID and Acute Coronary Syndromes: A State-of-the-art Review

  • Xiao Jiang1,#,
  • Yiran Lu2,#,
  • Yu Ding2,#,
  • Yuanyuan Liu3,
  • Zhen Zhao1,
  • Peizhong Liu1,
  • Chuangpeng Li1,
  • Song He2,
  • Qing Zhang2,
  • Rongyuan Yang1,*  and
  • Qing Liu1,* 
Exploratory Research and Hypothesis in Medicine   2025

doi: 10.14218/ERHM.2024.00043

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Jiang X, Lu Y, Ding Y, Liu Y, Zhao Z, Liu P, et al. Potential of Phytomedicine in Benefiting Both Long COVID and Acute Coronary Syndromes: A State-of-the-art Review. Explor Res Hypothesis Med. Published online: May 13, 2025. doi: 10.14218/ERHM.2024.00043.

Abstract

Acute coronary syndrome (ACS) in patients with SARS-CoV-2 infection is primarily driven by inflammation-induced myocardial injury through both direct and indirect mechanisms. Effective clinical management requires a dual approach: addressing cardiovascular lesions while also mitigating virus-induced local and systemic inflammation. This comprehensive approach is essential for improving the diagnosis and treatment of SARS-CoV-2-associated ACS. Emerging evidence highlights the potential of myocardial protective agents, including angiotensin-converting enzyme 2-modulating drugs and traditional Chinese medicine, which not only stabilize plaques and improve endothelial function but also confer cardioprotective effects. Furthermore, advancements in nanotechnology offer promising strategies for targeted therapy—particularly through angiotensin-converting enzyme 2 receptor modulation—by enhancing the precision and efficacy of herbal medicine delivery. This review explores the complex interplay between SARS-CoV-2 infection and ACS pathogenesis, and evaluates the therapeutic potential of pharmacological, herbal, and nanotechnology-based interventions in managing this multifaceted condition.

Keywords

Long COVID, Acute coronary syndromes, ACS, Phytomedicin, Nanotechnology, ACE2 receptor, Lipid nanoparticles

Introduction

Myocardial injury represents a severe comorbidity of SARS-CoV-2 infection, particularly when infection-triggered coronary occlusion precipitates acute coronary syndromes (ACS).1,2 As a critical cardiovascular manifestation, ACS substantially contributes to elevated mortality rates in affected populations.1,2 Emerging evidence has demonstrated that impaired ventilatory efficiency is a characteristic functional impairment in COVID-19 survivors developing ACS, highlighting persistent cardiopulmonary sequelae post-infection.3 However, the mechanisms by which viral infection triggers non-infectious ACS remain controversial. This mini-review aimed to highlight the potential association between long COVID and ACS while exploring the therapeutic potential of phytochemical compounds in alleviating SARS-CoV-2-related long-term symptoms in ACS patients.

Acute infection with SARS-CoV-2 triggers ACS

Acute infections, especially viral ones, may worsen ACS by triggering widespread inflammation that destabilizes plaques and impairs endothelial function.

In addition to the well-established view that ischemia in ACS is an aseptic inflammation process, acute infections by pathogens, including viruses and bacteria, are suggested to accelerate the pathological progression of ACS. Substantial elevations in serum inflammatory biomarkers—including acute-phase reactants (C-reactive protein and procalcitonin), neutrophil-derived enzymes (myeloperoxidase), and circulating leukocyte subsets (macrophages, T lymphocytes, and neutrophils)—are characteristically observed during ACS pathogenesis, with significantly higher levels than those detected in stable coronary artery disease patients.4,5

The inflammatory activity is not limited to the culprit lesion in the coronary artery but extends across the entire coronary tree, indicating that this is a systemic inflammation. Growing evidence suggests that acute infection-induced inflammatory responses may directly influence atherosclerotic plaque stability, coronary arterial integrity, and peri-adventitial adipose tissue through cellular mediators and their molecular effectors, including cytokine networks, proteolytic enzymes, coagulation cascade components, oxidative metabolites, and vascular tone regulators.5 These targeted inflammatory mechanisms potentiate endothelial dysfunction through vasa vasorum leakage, compromise fibrous cap integrity via necrotic core expansion, and propagate platelet-rich thrombus formation through coordinated P-selectin/GPIIb-IIIa interactions, collectively implicating sterile inflammation as a critical driver of both ACS initiation and progression.6

SARS-CoV-2 virus infection accelerates ACS through the angiotensin-converting enzyme (ACE) 2 receptor

SARS-CoV-2 induces heart damage via ACE2, causing inflammation, endothelial dysfunction, and clot formation that trigger myocardial injury and atherosclerosis through cytokine storms, plaque rupture, clotting changes, and oxygen supply issues, all of which increase the risk of ACS.

The acute SARS-CoV-2 infection led to a global pandemic, with acute myocardial injury (AMI) being prominently associated with comorbidities, alongside acute pulmonary injury.7 AMI is pathologically defined by substantial elevations in high-sensitivity cardiac troponin and/or brain natriuretic peptide, biomarkers that reflect myocardial stress from ischemic insults (e.g., plaque rupture-induced coronary occlusion) or non-ischemic etiologies such as ACS, decompensated heart failure, inflammatory cardiomyopathies, and arrhythmogenic myocardial remodeling.8

For ischemic heart disease, elevated troponin levels can be driven not only by direct coronary artery occlusion but also by indirect myocardial injury, such as the cardiovascular inflammation process, which may accelerate ischemic myocardial injury.9 Notably, the latter occurs during acute SARS-CoV-2 infection, when internalization of the SARS-CoV-2 virus requires the ACE2 receptor for transport.10,11

The extensive ACE2 expression in cardiovascular tissues predisposes them to SARS-CoV-2 infection, driving endothelial dysfunction and vascular inflammation. This cascade promotes thrombotic events, systemic inflammatory responses, and hypoxia, while adrenergic overactivation exacerbates myocardial stress during cytokine storm progression.12–14 These effects may contribute to triggering ACS and accelerating atherosclerosis. The mechanisms are as follows (Fig. 1):

COVID-19 invades the body through a large number of ACE2 receptors in the heart and blood vessels, affecting cytokine levels and triggering an inflammatory response.
Fig. 1  COVID-19 invades the body through a large number of ACE2 receptors in the heart and blood vessels, affecting cytokine levels and triggering an inflammatory response.

This leads to systemic cytokine storms, promotes thrombosis, and accelerates ACS. ACE2, angiotensin-converting enzyme 2; ACS, acute coronary syndrome; IL-6, interleukin-6.

  • Firstly, Elevated troponin levels correlate with inflammatory markers such as D-dimer (coagulation), ferritin (acute-phase response), interleukin (IL)-6 (cytokine activity), and lactate dehydrogenase (tissue injury), suggesting systemic immune dysregulation involving cytokine storms and reactive phagocytic/lymphocytic responses, rather than direct myocardial damage, during SARS-CoV-2 infection.1517

  • Secondly, marked inflammatory responses and hemodynamic instability predispose to atherosclerotic plaque rupture through mechanisms involving platelet activation/aggregation, ultimately precipitating acute coronary occlusion.18,19

  • Moreover, the inflammatory response impairs coronary endothelial function and hemostatic balance, increasing thrombin activity while reducing plasminogen activity,19 thus accelerating fibrin degradation products and D-dimer, leading to a pro-atherogenic and thrombogenic bias.20,21

  • Last but not least, the myocardial oxygen supply-demand imbalance due to hypoxia and tachycardia caused by systemic cytokine storms and pulmonary injury,22 probably becomes a synergistic factor that works with endothelial inflammation, vascular edema, and disseminated intravascular coagulation, ultimately exacerbating microvascular dysfunction, occlusion, or coronary spasm in myocardial tissue.2325

Thus, it is deduced that myocardial injury in patients with SARS-CoV-2 infection could be due to inflammation-induced direct endothelial and vascular injury, plaque rupture with platelet activation and aggregation, microthrombi formation, microvascular dysfunction and occlusion, or coronary spasm.

Myocardial protective drugs can regulate the ACE2 receptor in SARS-CoV-2 virus infection

COVID-19, though primarily a respiratory illness, also affects the heart, raising questions about the use of ACE inhibitors (ACEI) and angiotensin receptor blockers (ARB) due to their effects on ACE2. Meanwhile, melatonin shows promise in protecting the heart by improving blood vessel function and stabilizing plaques, potentially reducing ACS risk.

COVID-19 patients predominantly present with fever and cough, more frequently in adults than in children, along with dyspnea and myalgia, among other clinical features.26 As SARS-CoV-2 virus entry into cells is ACE2-dependent, and ACEI and ARB are widely used in cardiovascular disorders, the rationale for the use of ACEI or ARB needs to be considered.27,28 So far, there are conflicting data on whether these drugs increase or have minimal effects on ACE2 levels, which still require further investigation.

Nevertheless, reports on melatonin suggest a potential protective role against myocardial injury in SARS-CoV-2 infection. The oxygen supply-demand imbalance caused by infection-stimulated hemodynamic activation, hypoxemia, and inflammation-induced plaque instability with coronary hypoperfusion leads to ischemic heart damage. Melatonin could enhance the bioavailability of nitric oxide and improve coronary endothelial function, alleviating plaque instability by inhibiting intra-plaque inflammation. Thus, it exerts a myocardial protective function, potentially mitigating ACS risk and even myocardial ischemia-reperfusion injury.29 Notably, this does not imply that melatonin is a specific medication targeting acute inflammation in AMI, but it inspires exploration of novel therapeutic approaches for alleviating ACS injury in patients with acute inflammation, such as those with SARS-CoV-2 infection.

Herb benefits both long COVID and myocardial diseases

The COVID-19 pandemic has accelerated research into traditional Chinese medicine and herbal therapies, highlighting their dual antiviral and cardioprotective benefits. These agents work through multiple mechanisms—blocking viral entry, suppressing replication, and reducing inflammation—potentially lowering ACS risk in COVID-19 patients.

Since the first novel coronavirus pneumonia case in 2019, COVID-19 has become a worldwide infectious disease. Following the outbreak, long COVID became a common symptom in most populations. In China, more than 85% of patients received traditional Chinese herbal medicine therapy.30 Treatment with Chinese herbal medicine has been shown to relieve patients’ symptoms.31

Accordingly, the most commonly used herb for patients is licorice. While the antiviral mechanisms of glycyrrhizin remain incompletely characterized, emerging evidence suggests its interaction with multiple signaling pathways—including protein kinase C, casein kinase II, AP-1, p38 MAPK, and NF-κB—modulates DNA repair mechanisms and transcriptional regulation.32–34 Glycyrrhiza species exert cardioprotective effects via coordinated mechanisms, including attenuation of oxidative damage through Nrf2/ARE pathway activation, upregulation of endogenous antioxidant defenses, restoration of cardiac functional parameters, and preservation of myocardial structural integrity.35 Moreover, glycyrrhizic acid and its bioactive metabolite glycyrrhetinic acid suppress inducible nitric oxide synthase expression in activated macrophages, reducing nitric oxide synthesis—a critical driver of oxidative and inflammatory cascades in acute lung injury pathogenesis. Agastache rugosa, Forsythia suspensa, Atractylodes macrocephala, and Scutellaria baicalensis are also widely used in the mild and moderate stages of COVID-19.36 In clinical treatment, licorice is used in many traditional Chinese medicine prescriptions, such as QingFeiPaiDu decoction,37 ShuFengJieDu granules,38 JinHuaQingGan granules, and LianHua QingWen capsules.39

LianHua QingWen granules, a classic traditional Chinese medicine formula,40,41 have been approved to treat fever, fatigue, and other symptoms caused by mild COVID-19. Regarding the effect of herbs in the composition of LianHua QingWen granules, Chen et al.42 show that several anti-plague components from lotus play a potential role in inhibiting COVID-19 by significantly affecting the binding between ACE2 and S protein, which is a critical mechanism for preventing viral infection. Pan et al.43 showed that puerarin and quercetin may also combat COVID-19 through a similar mechanism by affecting the interaction between S protein and ACE2.

Astragalus membranaceus has a variety of pharmacological activities, including anti-virus, anti-inflammatory, and immune system regulation. It is rich in Astragalus polysaccharides, which have antiviral effects.44,45 Honeysuckle is also an effective anti-inflammatory Chinese herbal medicine, which is used for a variety of viral infections,46 such as hepatitis B virus,47 dengue virus,48 and intestinal and respiratory viruses, etc. These two herbs have been used since ancient China. Yeh et al.49 showed that honeysuckle-Astragalus preparation can not only improve the expression of a group of COVID-19-related miRNAs but also inhibit the expression of IL-6 and TNF-α, key inflammatory factors in cytokine storms, indicating that it has a certain inhibitory effect on cytokine storms. Furthermore, this preparation can inhibit the binding of COVID-19 protein to the ACE2 receptor, thus playing an antiviral role. In clinical practice, many Chinese patent medicines are in use, including Astragalus membranaceus or honeysuckle, such as Fuling Paidu decoction and Jinhua Qinggan capsules,50 which can be used to treat patients at various stages.51,52

Herb-derived medicine is also under investigation for the treatment of SARS-CoV-2 virus infection. These compounds or phytomedicines include Coronil,53 Cuphea ignea,54 Reynoutria Rhizomes,55 Cordycepin,56–58 Glycyrrhizic Acid,59–61 Perilla frutescens,62,63 Cyperus rotundus Linn,64 Thymoquinone,65–67 and others. The underlying mechanisms for the antiviral effects involve preventing the replication of the virus (e.g., Reynoutria Rhizomes),55 exhibiting inhibitory potential against the COVID-19 polymerase enzyme (RdRp) (e.g., Cordycepin),68 effectively killing the SARS-CoV-2 virus by targeting key protein structures of the virus or through immune synergy with other antiviral drugs (e.g., Glycyrrhizic Acid),59 blocking viral RNA and protein synthesis (e.g., Perilla frutescens leaf extract),62 inhibiting SARS-CoV-2 virus proteases to reduce viral replication, and antagonizing angiotensin-converting enzyme 2 receptors (e.g., Thymoquinone).69 Thus, these studies provide promising candidates for treating SARS-CoV-2 virus-induced ACS and, therefore, diminishing ACS injury (Table 1 and Fig. 2).32,40–45,49,50,54,57,63-69

Table 1

Mechanism of traditional Chinese medicine in the modulation of COVID-19 and myocardial diseases

NameCompositionPharmacological actionDrug target/related signalMechanism of actionMyocardial protectionStudy type (sample size)Population characteristicsKey findings (odds/hazard ratio)Reference
Honeysuckle Astragalus preparationAstragalus membranaceus, HoneysuckleAnti-virus, anti-inflammatory, regulating the immune systemIL-6, TNF-aSuppressing cytokine storm, inhibiting the binding of COVID-19 protein and ACE2 receptorYes65,66Review (one study); Preclinical (animal study); Pharmaceutical Analysis (two studies); Multiple Databases Analysis (one study)N/AN/A4045
LicoriceGlycyrrhizic AcidAnti-inflammatory, antioxidant, antitumorProtein kinase C, casein kinase, AP-1, MAPK-p38 and NF-κB, iNOSTargeting important protein structures of the virus or immune synergy effect with other antivirus drugs, Inhibition of acute lung injuryYes67Review (two studies)N/AN/A32,54
Nigella sativaThymoquinoneAntioxidant, anti-inflammatory, antiviral, antimicrobial, immunomodulatory, and anticoagulantCell surface heat shock protein (HSPA5), ACE2 receptor, IL-2, IL-4, IL-5, II-6, IL-12, IL-13Inhibiting virus proteases to diminish viral replication and as antagonism to angiotensin-converting ACE2 receptorsYes68Review (one study)N/AN/A64
Cuphea igneaAntiulcerogenic, antitumor, antioxidant and antihypertensiveInhibiting virus growthNoPharmaceutical Analysis (one study)N/AN/A49
Reynoutria RhizomesAntioxidant, antitumor, anti-inflammatory, and antiviralPreventing the replication of the virus, inhibiting virus growthNoPharmaceutical Analysis (one study)N/AN/A50
CordycepinAnti-inflammatory, antiviral, antibacterial, etc.Inhibiting RdRpYes69Molecular Dynamic simulation(one study)N/AN/A63
PerillaPerilla frutescens leaf extractLeaves are used to tonify stomach function, discharge heat, and improve healthy qi, and seeds decrease qi, resolve phlegm, relieve cough and asthma, and alleviate constipationCXCL10, IL-6, TNF-α, IFN-γ, MCP1Blocking viral RNA and protein synthesis, inhibiting proinflammatory factor expressionUnknownDrug Extraction(one study)N/AN/A57
Traditional Chinese medicine compounds inhibit viral invasion by acting on the ACE2 receptor, which is necessary for COVID-19 to enter the body.
Fig. 2  Traditional Chinese medicine compounds inhibit viral invasion by acting on the ACE2 receptor, which is necessary for COVID-19 to enter the body.

These compounds then affect downstream signaling pathways, such as ERK/MAPK, NF-κB, IRFs, and TGF-β, to reduce the expression of inflammatory factors, thereby aiding in the treatment of COVID-19. ACE2, angiotensin-converting enzyme 2; CHM, Chinese herbal medicine; EMT, Epithelial-Mesenchymal Transition; ERK, Extracellular Signal-Regulated Kinase; IFN-γ, interferon-γ; IL, interleukins; IRFS, Interferon regulatory factors; MAPK, Mitogen-Activated Protein Kinase; NF-κB, Nuclear Factor kappa-light-chain-enhancer of activated B cells; TGF-β, Transforming Growth Factor Beta; TNF-α, Tumor Necrosis Factor alpha.

Non-herbal medicine benefits both long COVID and myocardial diseases

The S proteins of SARS-CoV-1 and CoV-2 share 76% overall sequence identity, yet the receptor-binding domain of the latter has 10–20 times higher affinity for the human ACE2 receptor protein.70 After the receptor-binding domain binds ACE2, two heptad repeat domains, HR1 and HR2, interact to form a six-helical bundle, bringing the viral and host membranes close to one another, resulting in fusion. Using a recently solved crystal structure of the HR1 and HR2 domains of the SARS-CoV-2 S protein, lipidated peptide fusion inhibitors have been designed to inhibit pseudovirus infection of cells, with IC50 values in the single-digit nanomolar range.71 Due to its broad-spectrum anti-coronavirus activity, EK1C4 can be used for the treatment and prevention of infection not only by SARS-CoV-2 but also by other HCoVs. However, its fundamental limitation is the substantial development costs. Anakinra is the first recombinant IL-1 receptor antagonist that binds to both IL-1α and IL-1β receptors and has received approval from the U.S. Food and Drug Administration (FDA) for treating rheumatoid arthritis. In survival analysis, the development of any thromboembolic event, pulmonary thromboembolism, and ACS was higher in the SoC group compared to the Anakinra group. The survival rate was also lower in the SoC group than in the Anakinra group for patients who experienced thromboembolic events and ACS.72 However, its effectiveness in patients already suffering from respiratory failure has shown controversial results, and it is not recommended.73

Nanotechnology-based herbal medicine may benefit both SARS-CoV-2 virus infection and ACS by targeting the ACE2 receptor

Nanotechnology has revolutionized the COVID-19 response, enabling mRNA vaccine development and offering targeted therapies. Lipid nanoparticles have driven vaccine success, while nanoceria and silver nanoparticles show promise in fighting the virus and its complications through ACE2 modulation.

The U.S. FDA has granted Emergency Use Authorization for two mRNA vaccine candidates: Pfizer-BioNTech’s BNT162b2 and Moderna’s mRNA-1273.74 Emerging studies demonstrate that comprehensive targeting of conserved immunodominant epitopes across the full-length spike glycoprotein in SARS-CoV-2 variants, while harnessing the anti-inflammatory potential of regulatory T cells, provides critical insights for developing pan-coronavirus vaccines with broad-spectrum efficacy.75,76 The unprecedented efficacy of mRNA vaccines (BNT162b2 and mRNA-1273) has marked a turning point in pandemic containment. Nanoparticle platforms have proven instrumental in vaccine development, with lipid-based vectors shielding labile mRNA from ribonucleases while facilitating endosomal escape for cytosolic delivery. Phase III trials demonstrated 95% efficacy rates—a breakthrough stemming from innovative lipid nanoparticle delivery systems that optimize antigen presentation and enhance immunogenicity.77

Nanoceria (NC) is a rare-earth nano drug with catalase and superoxide dismutase mimic activity. In clinical studies, there is evidence that nanoceria can pass through TGF-β signaling pathways, potentially inhibiting the progression of fibrosis.78 NC may effectively inhibit fibrosis and reduce collagen deposition.79 Targeted transport of NC to the lungs may effectively alleviate acute respiratory distress syndrome.80

Targeted transportation may achieve better results. Silver nanoparticles (AgNPs) have also become one of the drugs that can inhibit SARS-CoV-2 virus infection due to their strong antiviral effect. AgNPs can attach to the virus genome to prevent viral replication and new virus release.81 AgNPs are likely to inhibit severe inflammatory responses, cytokine storms, and pulmonary fibrosis in COVID-19.82 More interestingly, reports have shown that cationic nanoparticles can directly bind ACE2, decrease its activity, and down-regulate its expression level in lung tissue, resulting in the deregulation of the renin-angiotensin system.83 Binding to ACE2 by multivalent attachment of ligands to nanocarriers incorporating antiviral therapeutics could increase receptor avidity and impart specificity to these nanovectors for host cells in the pulmonary tract. These findings suggest that nanoparticle-carrying herbal medicine may exert both cardioprotective effects in ACS and anti-inflammatory effects in SARS-CoV-2 virus infection by targeting the ACE2 receptor.

Nanotechnology plays a role in COVID-19 detection,84 diagnosis, treatment, and other stages.85 Nanobodies demonstrate therapeutic potential for COVID-19 by attenuating pulmonary hyperinflammation through targeted viral neutralization and immunomodulatory mechanisms.86 Nanodrugs have higher safety and biocompatibility and can provide more accurate drug targeting.87 However, nanoparticles also have limitations due to their toxicity. Nanoparticles have low solubility and degradability, so they can persist in cells for a long time and are not easily degraded.88 Furthermore, there is still a lack of a clear clearance mechanism.89 Therefore, the application of nanotechnology still needs to be further explored.

Future directions

Current research progress in phytomedicine has established critical directions for addressing SARS-CoV-2-associated cardiovascular complications, requiring systematic advancement across five key areas. First, large-scale, multicenter, randomized controlled trials should be prioritized to evaluate dose-response relationships of standardized phytochemical formulations (e.g., glycyrrhizic acid, astragaloside IV). These trials should be supported by longitudinal follow-up databases with a minimum three-year monitoring period to assess sustained efficacy and safety. Mechanistically, advanced structural biology techniques such as cryo-electron microscopy are needed to elucidate molecular interactions between bioactive plant compounds and ACE2 receptors. Concurrently, innovations in nanotechnology must address organ-targeting limitations through pharmacokinetic modeling to optimize nanoparticle delivery efficiency and biodegradability, alongside standardized toxicity assessments. Clinically, interdisciplinary frameworks should validate the synergistic effects between phytomedicine and conventional cardiovascular therapies across diverse ethnic populations, utilizing unified cardiovascular endpoints for efficacy evaluation. Furthermore, international consortia should integrate multi-omics platforms and leverage global COVID-19 cardiovascular sequelae registries to identify biomarkers predictive of phytotherapy responsiveness. These coordinated efforts will bridge gaps between preclinical research and clinical translation, ultimately informing evidence-based integrative cardiovascular care strategies.

Conclusions

Current research on SARS-CoV-2-associated ACS and herbal/nanotechnology therapies faces limitations, including reliance on observational data, unclear ACE2 mechanisms, and insufficient long-term safety evidence. Potential biases, such as overemphasis on positive results, may skew conclusions. SARS-CoV-2 triggers ACS through inflammation-induced myocardial injury, requiring therapies that target both cardiovascular damage and virus-driven inflammation. Promising approaches include ACE2-modulating drugs, traditional Chinese medicine, and nanotechnology, which stabilize plaques, protect endothelial function, and enable targeted delivery. However, challenges like nanoparticle toxicity must be addressed. A comprehensive, multi-modal strategy is essential for improving SARS-CoV-2-associated ACS outcomes. Compared to conventional therapeutic regimens, Chinese patent medicines demonstrate superior safety profiles characterized by a reduced incidence of adverse effects and lower treatment costs.

Declarations

Acknowledgement

None.

Funding

This study was supported by the Zhuhai Social Development Field Science and Technology Plan-Key Project (No. 2320004000286, to RYY) and the Basic and Applied Basic Research of Guangzhou City-University Joint Funding Project (No. 202201020382, to RYY).

Conflict of interest

The authors declare that there is no conflict of interest in the authorship and publication of this contribution.

Authors’ contributions

Contributed to study concept and design (QL, RYY, YD), drafting of the manuscript (XJ, YRL, HS,QZ), critical revision (YYL, ZZ), and finalization of the manuscript (QL). All authors read and approved the final manuscript (PZL, CPL).

References

  1. Timmis A, Vardas P, Townsend N, Torbica A, Katus H, De Smedt D, et al. European Society of Cardiology: cardiovascular disease statistics 2021: Executive Summary. European heart journal Quality of care & clinical outcomes 2022;8(4):377-382 View Article PubMed/NCBI
  2. GBD 2017 Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018;392(10159):1736-1788 View Article PubMed/NCBI
  3. Provenzale G, Barbieri L, Tumminello G, Carugo S, Guazzi M. Exercise response in post-acute coronary syndrome patients survived to COVID-19 infection. Int J Cardiol 2024;411:132285 View Article PubMed/NCBI
  4. Libby P, Hansson GK. Adaptive immunity in acute coronary syndromes: chicken or egg?. Eur Heart J 2018;39(13):1098-1099 View Article PubMed/NCBI
  5. Amsterdam EA, Wenger NK, Brindis RG, Casey DE, Ganiats TG, Holmes DR, et al. 2014 AHA/ACC Guideline for the Management of Patients with Non-ST-Elevation Acute Coronary Syndromes: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2014;64(24):e139-e228 View Article PubMed/NCBI
  6. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA 2020;323(11):1061-1069 View Article PubMed/NCBI
  7. Ayoubkhani D, Khunti K, Nafilyan V, Maddox T, Humberstone B, Diamond I, et al. Post-covid syndrome in individuals admitted to hospital with covid-19: retrospective cohort study. BMJ 2021;372:n693 View Article PubMed/NCBI
  8. Aye YN, Mai AS, Zhang A, Lim OZH, Lin N, Ng CH, et al. Acute myocardial infarction and myocarditis following COVID-19 vaccination. QJM 2023;116(4):279-283 View Article PubMed/NCBI
  9. Schmitz T, Meisinger C, Kirchberger I, Thilo C, Amann U, Baumeister SE, et al. Impact of COVID-19 pandemic lockdown on myocardial infarction care. Eur J Epidemiol 2021;36(6):619-627 View Article PubMed/NCBI
  10. Chen L, Li X, Chen M, Feng Y, Xiong C. The ACE2 expression in human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2. Cardiovasc Res 2020;116(6):1097-1100 View Article PubMed/NCBI
  11. Khan IH, Zahra SA, Zaim S, Harky A. At the heart of COVID-19. J Card Surg 2020;35(6):1287-1294 View Article PubMed/NCBI
  12. Alhogbani T. Acute myocarditis associated with novel Middle east respiratory syndrome coronavirus. Ann Saudi Med 2016;36(1):78-80 View Article PubMed/NCBI
  13. Zheng YY, Ma YT, Zhang JY, Xie X. COVID-19 and the cardiovascular system. Nat Rev Cardiol 2020;17(5):259-260 View Article PubMed/NCBI
  14. Imazio M, Klingel K, Kindermann I, Brucato A, De Rosa FG, Adler Y, et al. COVID-19 pandemic and troponin: indirect myocardial injury, myocardial inflammation or myocarditis?. Heart 2020;106(15):1127-1131 View Article PubMed/NCBI
  15. Jaffe AS, Cleland JGF, Katus HA. Myocardial injury in severe COVID-19 infection. Eur Heart J 2020;41(22):2080-2082 View Article PubMed/NCBI
  16. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395(10223):497-506 View Article PubMed/NCBI
  17. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 2020;395(10223):507-513 View Article PubMed/NCBI
  18. Driggin E, Madhavan MV, Bikdeli B, Chuich T, Laracy J, Biondi-Zoccai G, et al. Cardiovascular Considerations for Patients, Health Care Workers, and Health Systems During the COVID-19 Pandemic. J Am Coll Cardiol 2020;75(18):2352-2371 View Article PubMed/NCBI
  19. Libby P. Mechanisms of acute coronary syndromes and their implications for therapy. N Engl J Med 2013;368(21):2004-2013 View Article PubMed/NCBI
  20. Campello E, Bulato C, Spiezia L, Boscolo A, Poletto F, Cola M, et al. Thrombin generation in patients with COVID-19 with and without thromboprophylaxis. Clin Chem Lab Med 2021;59(7):1323-1330 View Article PubMed/NCBI
  21. Canzano P, Brambilla M, Porro B, Cosentino N, Tortorici E, Vicini S, et al. Platelet and Endothelial Activation as Potential Mechanisms Behind the Thrombotic Complications of COVID-19 Patients. JACC Basic Transl Sci 2021;6(3):202-218 View Article PubMed/NCBI
  22. Mahallawi WH, Khabour OF, Zhang Q, Makhdoum HM, Suliman BA. MERS-CoV infection in humans is associated with a pro-inflammatory Th1 and Th17 cytokine profile. Cytokine 2018;104:8-13 View Article PubMed/NCBI
  23. Wei ZY, Geng YJ, Huang J, Qian HY. Pathogenesis and management of myocardial injury in coronavirus disease 2019. Eur J Heart Fail 2020;22(11):1994-2006 View Article PubMed/NCBI
  24. Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature 2005;436(7047):112-116 View Article PubMed/NCBI
  25. Fu Y, Cheng Y, Wu Y. Understanding SARS-CoV-2-Mediated Inflammatory Responses: From Mechanisms to Potential Therapeutic Tools. Virol Sin 2020;35(3):266-271 View Article PubMed/NCBI
  26. Wang C, Horby PW, Hayden FG, Gao GF. A novel coronavirus outbreak of global health concern. Lancet 2020;395(10223):470-473 View Article PubMed/NCBI
  27. Kaseb AO, Mohamed YI, Malek AE, Raad II, Altameemi L, Li D, et al. The Impact of Angiotensin-Converting Enzyme 2 (ACE2) Expression on the Incidence and Severity of COVID-19 Infection. Pathogens 2021;10(3):379 View Article PubMed/NCBI
  28. Bojadzic D, Alcazar O, Chen J, Chuang ST, Condor Capcha JM, Shehadeh LA, et al. Small-Molecule Inhibitors of the Coronavirus Spike: ACE2 Protein-Protein Interaction as Blockers of Viral Attachment and Entry for SARS-CoV-2. ACS Infect Dis 2021;7(6):1519-1534 View Article PubMed/NCBI
  29. Fu Z, Jiao Y, Wang J, Zhang Y, Shen M, Reiter RJ, et al. Cardioprotective Role of Melatonin in Acute Myocardial Infarction. Front Physiol 2020;11:366 View Article PubMed/NCBI
  30. Ang L, Lee HW, Choi JY, Zhang J, Soo Lee M. Herbal medicine and pattern identification for treating COVID-19: a rapid review of guidelines. Integr Med Res 2020;9(2):100407 View Article PubMed/NCBI
  31. Yang Y, Islam MS, Wang J, Li Y, Chen X. Traditional Chinese Medicine in the Treatment of Patients Infected with 2019-New Coronavirus (SARS-CoV-2): A Review and Perspective. Int J Biol Sci 2020;16(10):1708-1717 View Article PubMed/NCBI
  32. Cinatl J, Morgenstern B, Bauer G, Chandra P, Rabenau H, Doerr HW. Glycyrrhizin, an active component of liquorice roots, and replication of SARS-associated coronavirus. Lancet 2003;361(9374):2045-2046 View Article PubMed/NCBI
  33. Chen F, Chan KH, Jiang Y, Kao RY, Lu HT, Fan KW, et al. In vitro susceptibility of 10 clinical isolates of SARS coronavirus to selected antiviral compounds. J Clin Virol 2004;31(1):69-75 View Article PubMed/NCBI
  34. Dos Santos WG. Natural history of COVID-19 and current knowledge on treatment therapeutic options. Biomed Pharmacother 2020;129:110493 View Article PubMed/NCBI
  35. Ojha SK, Sharma C, Golechha MJ, Bhatia J, Kumari S, Arya DS. Licorice treatment prevents oxidative stress, restores cardiac function, and salvages myocardium in rat model of myocardial injury. Toxicol Ind Health 2015;31(2):140-152 View Article PubMed/NCBI
  36. Gajewski A, Kośmider A, Nowacka A, Puk O, Wiciński M. Potential of herbal products in prevention and treatment of COVID-19. Literature review. Biomed Pharmacother 2021;143:112150 View Article PubMed/NCBI
  37. Yang R, Liu H, Bai C, Wang Y, Zhang X, Guo R, et al. Chemical composition and pharmacological mechanism of Qingfei Paidu Decoction and Ma Xing Shi Gan Decoction against Coronavirus Disease 2019 (COVID-19): In silico and experimental study. Pharmacol Res 2020;157:104820 View Article PubMed/NCBI
  38. Zhuang Z, Zhong X, Zhang H, Chen H, Huang B, Lin D, et al. Exploring the Potential Mechanism of Shufeng Jiedu Capsule for Treating COVID-19 by Comprehensive Network Pharmacological Approaches and Molecular Docking Validation. Comb Chem High Throughput Screen 2021;24(9):1377-1394 View Article PubMed/NCBI
  39. Xia QD, Xun Y, Lu JL, Lu YC, Yang YY, Zhou P, et al. Network pharmacology and molecular docking analyses on Lianhua Qingwen capsule indicate Akt1 is a potential target to treat and prevent COVID-19. Cell Prolif 2020;53(12):e12949 View Article PubMed/NCBI
  40. Runfeng L, Yunlong H, Jicheng H, Weiqi P, Qinhai M, Yongxia S, et al. Lianhuaqingwen exerts anti-viral and anti-inflammatory activity against novel coronavirus (SARS-CoV-2). Pharmacol Res 2020;156:104761 View Article PubMed/NCBI
  41. Hu K, Guan WJ, Bi Y, Zhang W, Li L, Zhang B, et al. Efficacy and safety of Lianhuaqingwen capsules, a repurposed Chinese herb, in patients with coronavirus disease 2019: A multicenter, prospective, randomized controlled trial. Phytomedicine 2021;85:153242 View Article PubMed/NCBI
  42. Chen X, Wu Y, Chen C, Gu Y, Zhu C, Wang S, et al. Identifying potential anti-COVID-19 pharmacological components of traditional Chinese medicine Lianhuaqingwen capsule based on human exposure and ACE2 biochromatography screening. Acta Pharm Sin B 2021;11(1):222-236 View Article PubMed/NCBI
  43. Pan B, Fang S, Zhang J, Pan Y, Liu H, Wang Y, et al. Chinese herbal compounds against SARS-CoV-2: Puerarin and quercetin impair the binding of viral S-protein to ACE2 receptor. Comput Struct Biotechnol J 2020;18:3518-3527 View Article PubMed/NCBI
  44. Zheng Y, Ren W, Zhang L, Zhang Y, Liu D, Liu Y. A Review of the Pharmacological Action of Astragalus Polysaccharide. Front Pharmacol 2020;11:349 View Article PubMed/NCBI
  45. Xue H, Gan F, Zhang Z, Hu J, Chen X, Huang K. Astragalus polysaccharides inhibits PCV2 replication by inhibiting oxidative stress and blocking NF-κB pathway. Int J Biol Macromol 2015;81:22-30 View Article PubMed/NCBI
  46. Yu Y, Zhu C, Wang S, Song W, Yang Y, Shi J. Homosecoiridoid alkaloids with amino acid units from the flower buds of Lonicera japonica. J Nat Prod 2013;76(12):2226-2233 View Article PubMed/NCBI
  47. Ge L, Xiao L, Wan H, Li J, Lv K, Peng S, et al. Chemical constituents from Lonicera japonica flower buds and their anti-hepatoma and anti-HBV activities. Bioorg Chem 2019;92:103198 View Article PubMed/NCBI
  48. Lee YR, Yeh SF, Ruan XM, Zhang H, Hsu SD, Huang HD, et al. Honeysuckle aqueous extract and induced let-7a suppress dengue virus type 2 replication and pathogenesis. J Ethnopharmacol 2017;198:109-121 View Article PubMed/NCBI
  49. Yeh YC, Doan LH, Huang ZY, Chu LW, Shi TH, Lee YR, et al. Honeysuckle (Lonicera japonica) and Huangqi (Astragalus membranaceus) Suppress SARS-CoV-2 Entry and COVID-19 Related Cytokine Storm in Vitro. Front Pharmacol 2021;12:765553 View Article PubMed/NCBI
  50. Liu Z, Li X, Gou C, Li L, Luo X, Zhang C, et al. Effect of Jinhua Qinggan granules on novel coronavirus pneumonia in patients. J Tradit Chin Med 2020;40(3):467-472 View Article PubMed/NCBI
  51. Lee DYW, Li QY, Liu J, Efferth T. Traditional Chinese herbal medicine at the forefront battle against COVID-19: Clinical experience and scientific basis. Phytomedicine 2021;80:153337 View Article PubMed/NCBI
  52. Zang Y, Wan J, Zhang Z, Huang S, Liu X, Zhang W. An updated role of astragaloside IV in heart failure. Biomed Pharmacother 2020;126:110012 View Article PubMed/NCBI
  53. Balkrishna A, Tomer M, Verma S, Joshi M, Sharma P, Srivastava J, et al. Phytometabolite profiling of Coronil, a herbal medicine for COVID-19, its identification by mass-spectroscopy and quality validation on liquid chromatographic platforms. J Sep Sci 2021;44(22):4064-4081 View Article PubMed/NCBI
  54. Mahmoud DB, Ismail WM, Moatasim Y, Kutkat O, ElMeshad AN, Ezzat SM, et al. Delineating a potent antiviral activity of Cuphea ignea extract loaded nano-formulation against SARS-CoV-2: In silico and in vitro studies. J Drug Deliv Sci Technol 2021;66:102845 View Article PubMed/NCBI
  55. Nawrot-Hadzik I, Zmudzinski M, Matkowski A, Preissner R, Kęsik-Brodacka M, Hadzik J, et al. Reynoutria Rhizomes as a Natural Source of SARS-CoV-2 Mpro Inhibitors-Molecular Docking and In Vitro Study. Pharmaceuticals (Basel) 2021;14(8):742 View Article PubMed/NCBI
  56. Verma AK. Cordycepin: a bioactive metabolite of Cordyceps militaris and polyadenylation inhibitor with therapeutic potential against COVID-19. J Biomol Struct Dyn 2022;40(8):3745-3752 View Article PubMed/NCBI
  57. Verma AK, Aggarwal R. Repurposing potential of FDA-approved and investigational drugs for COVID-19 targeting SARS-CoV-2 spike and main protease and validation by machine learning algorithm. Chem Biol Drug Des 2021;97(4):836-853 View Article PubMed/NCBI
  58. Rabie AM. Potent Inhibitory Activities of the Adenosine Analogue Cordycepin on SARS-CoV-2 Replication. ACS Omega 2022;7(3):2960-2969 View Article PubMed/NCBI
  59. Sun Z, He G, Huang N, Thilakavathy K, Lim JCW, Kumar SS, et al. Glycyrrhizic Acid: A Natural Plant Ingredient as a Drug Candidate to Treat COVID-19. Front Pharmacol 2021;12:707205 View Article PubMed/NCBI
  60. Zheng W, Huang X, Lai Y, Liu X, Jiang Y, Zhan S. Glycyrrhizic Acid for COVID-19: Findings of Targeting Pivotal Inflammatory Pathways Triggered by SARS-CoV-2. Front Pharmacol 2021;12:631206 View Article PubMed/NCBI
  61. Yu S, Zhu Y, Xu J, Yao G, Zhang P, Wang M, et al. Glycyrrhizic acid exerts inhibitory activity against the spike protein of SARS-CoV-2. Phytomedicine 2021;85:153364 View Article PubMed/NCBI
  62. Tang WF, Tsai HP, Chang YH, Chang TY, Hsieh CF, Lin CY, et al. Perilla (Perilla frutescens) leaf extract inhibits SARS-CoV-2 via direct virus inactivation. Biomed J 2021;44(3):293-303 View Article PubMed/NCBI
  63. Jan JT, Cheng TR, Juang YP, Ma HH, Wu YT, Yang WB, et al. Identification of existing pharmaceuticals and herbal medicines as inhibitors of SARS-CoV-2 infection. Proc Natl Acad Sci U S A 2021;118(5):e2021579118 View Article PubMed/NCBI
  64. Kumar SB, Krishna S, Pradeep S, Mathews DE, Pattabiraman R, Murahari M, et al. Screening of natural compounds from Cyperus rotundus Linn against SARS-CoV-2 main protease (M(pro)): An integrated computational approach. Comput Biol Med 2021;134:104524 View Article PubMed/NCBI
  65. Ahmad A, Raish M, Alkharfy KM. The potential role of thymoquinone in preventing the cardiovascular complications of COVID-19. Vascul Pharmacol 2021;141:106899 View Article PubMed/NCBI
  66. Xu H, Liu B, Xiao Z, Zhou M, Ge L, Jia F, et al. Computational and Experimental Studies Reveal That Thymoquinone Blocks the Entry of Coronaviruses Into In Vitro Cells. Infect Dis Ther 2021;10(1):483-494 View Article PubMed/NCBI
  67. Esharkawy ER, Almalki F, Hadda TB. In vitro potential antiviral SARS-CoV-19- activity of natural product thymohydroquinone and dithymoquinone from Nigella sativa. Bioorg Chem 2022;120:105587 View Article PubMed/NCBI
  68. Bibi S, Hasan MM, Wang YB, Papadakos SP, Yu H. Cordycepin as a Promising Inhibitor of SARS-CoV-2 RNA Dependent RNA Polymerase (RdRp). Curr Med Chem 2022;29(1):152-162 View Article PubMed/NCBI
  69. Badary OA, Hamza MS, Tikamdas R. Thymoquinone: A Promising Natural Compound with Potential Benefits for COVID-19 Prevention and Cure. Drug Des Devel Ther 2021;15:1819-1833 View Article PubMed/NCBI
  70. Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, et al. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science 2020;367(6483):1260-1263 View Article PubMed/NCBI
  71. Xia S, Liu M, Wang C, Xu W, Lan Q, Feng S, et al. Inhibition of SARS-CoV-2 (previously 2019-nCoV) infection by a highly potent pan-coronavirus fusion inhibitor targeting its spike protein that harbors a high capacity to mediate membrane fusion. Cell Res 2020;30(4):343-355 View Article PubMed/NCBI
  72. Çakmak R, Yüce S, Ay M, Uyar MH, Kılıç Mİ, Bektaş M. Intravenous high-dose anakinra drops venous thrombosis and acute coronary syndrome in severe and critical COVID-19 patients: a propensity score matched study. Sci Rep 2024;14(1):12369 View Article PubMed/NCBI
  73. Khani E, Shahrabi M, Rezaei H, Pourkarim F, Afsharirad H, Solduzian M. Current evidence on the use of anakinra in COVID-19. Int Immunopharmacol 2022;111:109075 View Article PubMed/NCBI
  74. Meo SA, Bukhari IA, Akram J, Meo AS, Klonoff DC. COVID-19 vaccines: comparison of biological, pharmacological characteristics and adverse effects of Pfizer/BioNTech and Moderna Vaccines. Eur Rev Med Pharmacol Sci 2021;25(3):1663-1669 View Article PubMed/NCBI
  75. Kumar R, Srivastava Y, Muthuramalingam P, Singh SK, Verma G, Tiwari S, et al. Understanding Mutations in Human SARS-CoV-2 Spike Glycoprotein: A Systematic Review & Meta-Analysis. Viruses 2023;15(4):856 View Article PubMed/NCBI
  76. Dhawan M, Rabaan AA, Alwarthan S, Alhajri M, Halwani MA, Alshengeti A, et al. Regulatory T Cells (Tregs) and COVID-19: Unveiling the Mechanisms, and Therapeutic Potentialities with a Special Focus on Long COVID. Vaccines (Basel) 2023;11(3):699 View Article PubMed/NCBI
  77. Khurana A, Allawadhi P, Khurana I, Allwadhi S, Weiskirchen R, Banothu AK, et al. Role of nanotechnology behind the success of mRNA vaccines for COVID-19. Nano Today 2021;38:101142 View Article PubMed/NCBI
  78. Lai CC, Shih TP, Ko WC, Tang HJ, Hsueh PR. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): The epidemic and the challenges. Int J Antimicrob Agents 2020;55(3):105924 View Article PubMed/NCBI
  79. Oró D, Yudina T, Fernández-Varo G, Casals E, Reichenbach V, Casals G, et al. Cerium oxide nanoparticles reduce steatosis, portal hypertension and display anti-inflammatory properties in rats with liver fibrosis. J Hepatol 2016;64(3):691-698 View Article PubMed/NCBI
  80. Allawadhi P, Khurana A, Allwadhi S, Joshi K, Packirisamy G, Bharani KK. Nanoceria as a possible agent for the management of COVID-19. Nano Today 2020;35:100982 View Article PubMed/NCBI
  81. Salleh A, Naomi R, Utami ND, Mohammad AW, Mahmoudi E, Mustafa N, et al. The Potential of Silver Nanoparticles for Antiviral and Antibacterial Applications: A Mechanism of Action. Nanomaterials (Basel) 2020;10(8):1566 View Article PubMed/NCBI
  82. Allawadhi P, Singh V, Khurana A, Khurana I, Allwadhi S, Kumar P, et al. Silver nanoparticle based multifunctional approach for combating COVID-19. Sens Int 2021;2:100101 View Article PubMed/NCBI
  83. Deloukas P, Kanoni S, Willenborg C, Farrall M, Assimes TL, Thompson JR, et al. Large-scale association analysis identifies new risk loci for coronary artery disease. Nat Genet 2013;45(1):25-33 View Article PubMed/NCBI
  84. Alhalaili B, Popescu IN, Kamoun O, Alzubi F, Alawadhia S, Vidu R. Nanobiosensors for the Detection of Novel Coronavirus 2019-nCoV and Other Pandemic/Epidemic Respiratory Viruses: A Review. Sensors (Basel) 2020;20(22):6591 View Article PubMed/NCBI
  85. Rai M, Bonde S, Yadav A, Bhowmik A, Rathod S, Ingle P, et al. Nanotechnology as a Shield against COVID-19: Current Advancement and Limitations. Viruses 2021;13(7):1224 View Article PubMed/NCBI
  86. Verma G, Dhawan M, Saied AA, Kaur G, Kumar R, Emran TB. Immunomodulatory approaches in managing lung inflammation in COVID-19: A double-edge sword. Immun Inflamm Dis 2023;11(9):e1020 View Article PubMed/NCBI
  87. Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol 2010;10(11):787-796 View Article PubMed/NCBI
  88. Kim HP, Kim JK, Jo MS, Park JD, Ahn K, Gulumian M, et al. Even lobar deposition of poorly soluble gold nanoparticles (AuNPs) is similar to that of soluble silver nanoparticles (AgNPs). Part Fibre Toxicol 2020;17(1):54 View Article PubMed/NCBI
  89. Osman NM, Sexton DW, Saleem IY. Toxicological assessment of nanoparticle interactions with the pulmonary system. Nanotoxicology 2020;14(1):21-58 View Article PubMed/NCBI

About this Article

Cite this article
Jiang X, Lu Y, Ding Y, Liu Y, Zhao Z, Liu P, et al. Potential of Phytomedicine in Benefiting Both Long COVID and Acute Coronary Syndromes: A State-of-the-art Review. Explor Res Hypothesis Med. Published online: May 13, 2025. doi: 10.14218/ERHM.2024.00043.
Copy Export to RIS Export to EndNote
Article History
Received Revised Accepted Published
December 5, 2024 April 2, 2025 April 10, 2025 May 13, 2025
DOI http://dx.doi.org/10.14218/ERHM.2024.00043