Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Med., 23 September 2025

Sec. Infectious Diseases: Pathogenesis and Therapy

Volume 12 - 2025 | https://doi.org/10.3389/fmed.2025.1569013

Comprehensive review on COVID-19: etiology, pathogenicity, and treatment


Amr El Zawily
Amr El Zawily1*Sarah EckertSarah Eckert2Reed AdajarReed Adajar2Nardin WagihNardin Wagih3Abeer H. ElmaidomyAbeer H. Elmaidomy4Abdelrahman M. HelmyAbdelrahman M. Helmy5Mazen MustafaMazen Mustafa6Mazhar ElshorbagiMazhar Elshorbagi7Erin GhaliErin Ghali8Rehab G. FadlRehab G. Fadl8Jochen BodemJochen Bodem9Usama Ramadan Abdelmohsen,
Usama Ramadan Abdelmohsen6,10*Marco Y. W. Zaki,Marco Y. W. Zaki8,11
  • 1University of Guelph, Department of Molecular and Cellular Biology, Guelph, ON, Canada
  • 2Department of Biology, University of Iowa, Iowa City, IA, United States
  • 3Department of Medicinal Chemistry, Faculty of Pharmacy, Deraya University, Minia, Egypt
  • 4Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
  • 5Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Badr University in Assiut, Assiut, Egypt
  • 6Deraya Center for Scientific Research, Deraya University, Minia, Egypt
  • 7Department of Philosophy, Deraya University, Minia, Egypt
  • 8Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
  • 9Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
  • 10Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, Egypt
  • 11Centre for Research and Sustainability, Deraya University, Minia, Egypt

With the unprecedented surge of severe COVID-19 cases in early 2020, researchers and medical professionals worked actively to identify effective viral infection treatments based on a scientific understanding of viruses. Over the past few years, an enormous amount of research has investigated the viral infection and replication processes following the first SARS-CoV-2 case. With this knowledge, many drugs have been explicitly created to inhibit viral replication or decrease the severity of the immune response. Additionally, scientists have utilized decades of research and techniques to expedite SARS-CoV-2 vaccine development. SARS-CoV-2, a positive-strand RNA virus, belongs to the Sarbecovirus subgroup of Betacoronaviruses. Its emergence is not unique; previous outbreaks like SARS and MERS have shaped our understanding of coronavirus-related diseases. Molecular clock analysis suggests that the ancestor of all current coronaviruses existed over 10,000 years ago, with subsequent evolution occurring around 3300–2400 BC. Researchers have explored synthetic and natural treatments alongside other antiviral therapies, corticosteroids, and immunotherapies. Additionally, using artificial intelligence and nano-based technologies enriched SARS-CoV-2 diagnosis and management. In this comprehensive review, we provide recent literature on COVID-19, exploring its evolving etiology, pathogenicity, and pathophysiology, alongside developments in synthetic and natural therapeutic strategies, vaccines, artificial intelligence in diagnosis, and nano-based technologies.

Introduction

Coronavirus is a positive-strand RNA virus that infects many mammalian species, causing respiratory tract-related diseases of varying severity (1). The SARS-CoV-2 virus that caused the latest outbreak is a member of the Sarbecovirus subgroup of the Betacoronavirus (1). The evolution of the severe acute respiratory syndrome (SARS) caused by SARS-CoV-1 in 2003 and the Middle East Respiratory Syndrome (MERS) caused by MERS-CoV in 2012 preceded the outburst of the current SARS-CoV-2 pandemic (2). Other milder or sub-clinical coronavirus-associated diseases further support the idea that SARS-CoV-2 was not extraordinary and may reappear in the future (3). Molecular clock analysis revealed that the ancestor of all the current groups of coronaviruses existed more than 10,000 years ago, with different groups of coronaviruses evolving around 3300–2400 BC (1). The rapid SARS-CoV-2 transmission and the uncertainty of its related effects rushed the introduction of new treatment regimens to contain the pandemic. The review outlines the virus's etiology, pathogenicity, and pathophysiology. It details various synthetic and natural therapies exploring their efficacy in preventing or treating COVID-19 based on the available in-silico, in-vitro, in-vivo, and clinical data (Table 1). Finally, different types of SARS-CoV-2 vaccines, their effectiveness, safety, and potential future improvements, including artificial intelligence advancements and nano-based technologies, are thoroughly addressed.

Table 1
www.frontiersin.org

Table 1. Comparison of COVID-19 vaccine platforms: mechanisms, advantages, limitations, and clinical status.

Evolution of SARS-CoV-2

The origin of SARS-CoV-2

Most coronavirus strains are animal-derived. However, host coinfection with different variants of SARS-related coronaviruses can give rise to viral recombination, which is one hypothesis of how SARS-CoV-2 has evolved (4). The phylogenetic relativity between the SARS-CoV-2 and Yunnan's RaTG13 bat virus is noteworthy; the sequence similarity between both strains is around 96% (5). However, synonymous substitutions, which do not alter the amino acid sequence—between both strains reduced that similarity to only 83% (6). The error rate in coronavirus replication is significantly lower than that of influenza due to the viral proofreading exonuclease nsp14 (7, 8), and the synonymous evolutionary rate of SARS-CoV-2 ranges between 1.19–1.31 × 10−3/site/year (913). This leads to a divergence time between SARS-CoV-2 and RaTG13, between 18 and 71.4 years (12, 13). The advanced Bayesian phylogenetic method hypothesized that SARS-CoV-2 and RaTG13 did exist with their Most Recent Common Ancestor (MRCA) in 1969 (14), suggesting that both SARS-CoV-2 and RaTG13 might be divergent and pointing to the need to find other coronaviruses that are more similar to SARS-CoV-2 than RaTG13. The alignment of 68 different Sarbecovirus strains revealed multiple “recombination blocks” between recombination breakpoints in the SARS-CoV-2 genome, especially in areas translated to ORF1 and the S protein (14). Specifically, the ORF1b, the 5′ region of the S protein, and the nucleocapsid protein regions in both SARS-CoV-2 and RaTG13 showed more similarity than other coronavirus species (14). Interestingly, in these genomic regions, the 2019 Pangolin Guangdong coronavirus (15) showed a solid relationship to the ancestor of both SARS-CoV-2 and RaTG13 (14, 15). Strikingly, the SARS-CoV-2 variable loop region of the S protein, which contains six residues in the Angiotensin Converting Enzyme 2 (ACE-2) Receptor Binding Domain (RBD), had more substantial similarity to the sequence of Pangolin Guangdong coronavirus compared to that of RaTG13 (15). This either suggests a potential recombination between SARS-CoV-2 and the Pangolin Guangdong strain after the split of SARS-CoV-2 and RaTG13 lineages or a recombination between Pangolin Guangdong and the common ancestral strain of both SARS-CoV-2 and RaTG13 with the later gaining more sequence changes from the SARS-CoV-2 strain (14, 15). Phylogenetic analysis of the S protein also showed higher mutability for deletions, mutations, and recombination (16) in line with other studies that confirmed the divergence of the RBD region in the S protein of SARS-CoV-2 from the RaTG13 (17). Although it is hard to determine whether the evolution of SARS-CoV-2 occurred via direct transmission or an intermediate host, the evolution of the SARS-CoV-2 genome witnessed a series of recombination events from other viruses. Altogether, there are different tiers of complexity in the structure and evolution of the S protein, among other parts of the SARS-CoV-2 genome. Further investigation into the functional relevance of these events to the infection and progression process is warranted.

SARS-CoV-2 variants

The evolution of new SARS-CoV-2 variants can be attributed to point mutations, recombination between SARS-CoV-2 variants, and host-mediated RNA editing through the Apolipoprotein B mRNA Editing enzyme, Catalytic polypeptide (APOBEC) and Adenosine Deaminase Acting on RNA (ADAR) enzymes (14). Emergent SARS-CoV-2 variants are hard to trace and classify due to low nucleotide substitutions and the lack of appropriate sequencing techniques in some countries witnessing their outbreak (18). The substitution of one amino acid residue (D614G) in the spike protein from the Wuhan-Hu-1 sequence (NC_045512.2) became 100% prevalent by June 2020. It was the first evidence of the SARS-CoV-2 evolutionary selective pressure (19). At the end of 2020, the UK reported that infection of the new Alpha (B.1.1.7) variant was first reported in September of the same year. This fast-rising lineage was associated with a higher rate of mutagenesis of its spike proteins. A few weeks later, South Africa reported the evolution of the Beta (B.1.351) variant that regionally predominated in just 2 months (10). In Brazil, the Gamma (P.1) variant occurred with a prevalence of 75% by October 2020 (20). In 2021, another variant, Delta (B.1.617.2), was reported in India and spread to many other countries (21). The transmission rate and the severity of the B.1.617.2 variant surpassed earlier virus variants (21). Given these genetic, epidemiological, transmission, and immune evading disparities, the World Health Organization (WHO), COVID-19 Genomic UK consortium, and the US Centres for Disease Control and Prevention (CDC) have classified the SARS-CoV-2 lineages into Variants Of Concern (VOCs) and Variants Of Interest (VOIs) (22, 23). VOCs are variants that induce more severe diseases with higher transmission rates and immune evasion potential. They were further subclassified into Alpha, Beta, Gamma, and Delta (Table 2). Less common variants, albeit with a similar mutational burden to the VOCs, were categorized under the “VOIs” classification (22, 23). The Omicron variant, which emerged in late 2021, infected primary adult upper airway tissue relative to Delta, resulting in higher transmissibility (24, 25).

Table 2
www.frontiersin.org

Table 2. Recent evolution of SARS-CoV-2 variants (50, 300302).

The Omicron family of SARS-CoV-2 sub-variants represents the most genetically diverse and rapidly evolving lineage of the virus since its emergence in late 2021. The progenitor variant, B.1.1.529 (Omicron), was initially identified in South Africa. It exhibited over 50 mutations on the spike protein, which enhanced its transmissibility and increased its ability to evade immune responses (19).

Throughout 2022, sub-lineages including BA.1, BA.2, and BA.5 quickly replaced the Delta form, which was previously the most prevalent. As the virus developed further, novel sub-variants including EG.5 (Eris), JN.1, and XBB.1.5 (Kraken) appeared. These sub-variants all demonstrated additional adaptations in immune evasion and infection rates while generally retaining milder clinical presentations, particularly in vaccinated populations (19).

By 2024–2025, JN.1 had been the most common type globally. This was followed by the appearance of NB.1.8.1, which caused concern because of its quick expansion in China and its discovery in foreign visitors. Derived from JN.1, NB.1.8.1 and other more recent sub-lineages carry on the pattern of increased transmissibility and partial resistance to neutralizing antibodies. A runny nose, fatigue, fever, headaches, coughing, sore throat, dyspnoea, digestive problems, and loss of taste and smell are some of the symptoms (19). Table 2 outlines an up-to-date summary of the original prevalent and dangerous variants, and the newest prevalent variants that are either of interest or of concern.

Etiology

The main risk factors of SARS-CoV-2 infection are male gender, older age, lower socioeconomic status, ethnicity, chronic disease, and country of birth [reviewed in ref. (26)]. It is essential to recognize that COVID-19 is an etiology to many diseases, such as cold agglutinin disease (CAD), autoimmune hemolytic anemia (AIHA), acute kidney injury (AKI), and more (27, 28). Identifying and treating these co-factors during viral infection may improve patient recovery. Many studies have shown that pre-existing conditions are risk factors for contracting COVID-19 and exacerbate symptoms during viral infection. Age and diabetes correlated with severe symptomology. Numbers were even higher when diabetes coexisted with comorbidities like hypertension (29, 30). Cardiovascular, respiratory, or hypertension comorbidities, those requiring ventilation, and those with elevated sequential organ failure assessment (SOFA) scores also have a higher likelihood not only of hospitalization in the ICU with acute respiratory distress syndrome (ARDS) but also a higher risk of death when contracting COVID-19 (31). A case study from early 2020 found that patients with bacterial or fungal co-infections were more likely to experience complications (32). Epilepsy appears to be more commonly associated with an increased risk of neurological manifestations and abnormal electrical activity in the brain when diagnosed with COVID-19 (33). Infected patients with chronic kidney disease often have the most impaired immune response out of patients with other comorbidities (34).

SARS-CoV-2 serves as an etiology itself to numerous inflammatory responses and disorders. Infected persons commonly experience cough, fatigue, and fever, although asymptomatic conditions may occur (35). The virus is known to lead to neurological syndromes, such as stroke, coma, anosmia, headache, and ageusia (35). Infections can directly affect glomeruli and renal tubules, cell-mediated immunity, and cytokine storms (36). Five to twenty-three percent of people with the coronavirus develop symptoms of acute kidney injury (AKI), which coincides with the activation of coagulation pathways, damage to the renal vascular endothelium, increased cytokines and cytokine storm, sepsis, circulatory disturbances, and hypoxemia (28). Numerous case studies show that foot manifestations (intense pain, numbness, itching, swelling, rashes, and/or altered gait) are symptoms of COVID-19 infection (37). Additionally, infection has been shown to damage the cardiovascular system, causing myocardial infarction, heart failure, cardiogenic shock, myocarditis, microthrombi, and stress cardiomyopathy (38).

Pathogenesis of SARS-CoV-2

A typical SARS-CoV-2 infection involves viral invasion and multiplication, dysregulated immune response, multiple organ damage, and recovery (39). As the virus penetrates the host cells, it replicates, assembles, and most likely affects alveolar epithelial cells, leading to bilateral diffuse alveolar damage, vascular obstruction, patchy inflammatory cellular infiltration, intra-alveolar edema, hemorrhage, proteinaceous exudate, denudation, and reactive pneumocyte hyperplasia (40, 41). ARDS, sepsis, and multiple organ dysfunction syndrome (MODS) are all caused by the simultaneous release of numerous Pathogen-Associated Molecular Patterns (PAMPs) and Damage-Associated Molecular Patterns (DAMPs), eliciting immune cell infiltration. Although most patients eventually recover, some may experience chronic disease, persistent inflammation, systemic immunosuppression, and development of catabolic syndrome (42).

Host cell invasion by SARS-CoV-2

SARS-CoV-2 has four main structural proteins that contribute to viral infection. The spike (S) protein mediates the binding of the virus to the ACE2 receptor on host cells, the envelope (E) protein activates immunopathology in infection, the membrane (M) protein stabilizes the viral structure and its budding and release, and the nucleocapsid (N) protein takes part in viral replication (43). Three non-structural viral proteins play a critical role in viral replication and the immune response: RNA-dependent RNA polymerase (RdRp), main protease (Mpro), and papain-like protease (PLpro) (44) (Figure 1). RdRp plays a significant role in the replication of -ssRNA viruses, and Mpro and PLpro are cysteine proteases that process the polyproteins (45).

Figure 1
Diagram illustrating the SARS-CoV-) replication cycle and therapeutic targets. Panel A shows virus entry via cell surface and endosomal routes, with inhibitors like hrsACE) and Chloroquine depicted. Panel B details viral RNA translation, transcription, and replication with highlighted antiviral drugs such as Remdesivir and Favipiravir. Panel C outlines virion assembly and release through the rough ER, Golgi, and exocytosis. The graphic emphasizes intervention points for potential treatments with labeled compounds and enzymes.

Figure 1. SARS-COV-2 life cycle and therapeutic targets. (A) Attachment and entry: SARS-CoV-2 can enter host cells either via endosomes or via fusion between the viral envelope and the host cell membrane. Both routes involve viral S protein binding to the entry receptor ACE2 on host cell, triggering conformational changes in S1 subunit and exposing the S2′ cleavage site in the S2 subunit. In the presence of TMPRSS2, it cleaves the S2′ site directly at the cell surface (cell surface entry). While in the absence of TMPRSS2 (endosomal entry), the virus–ACE2 complex is taken into the cell through clathrin-mediated endocytosis. consequently, cathepsins in acidic endolysosomes cleave the S2′ site. Fusion initiates as S1 dissociates from S2, inducing major conformational changes in the S2 subunit, and leading to protrusion of the FP forward into the target membrane OR as the cleavage of the S2′ site exposes the fusion peptide (FP). Viral RNA is released into the cytoplasm after formation of fusion pores. (1) Agents that disrupt interaction between the S protein and ACE2 receptor can inhibit both entry pathways. Ex: human recombinant soluble ACE2 (hrsACE2), an ACE2 mimetic, have been shown to inhibit the attachment of the virus to the cells by misleading viruses to bind to a pseudo receptor, and antibodies released by vaccination block virus binding to ACE2. Serine protease inhibitors camostat mesylate and nafamostat restrict the TMPRSS2-mediated entry pathway. Antimalarials hydroxychloroquine and chloroquine block endosomal acidification, which is necessary for cathepsin activity, consequently limiting the cathepsin-mediated entry pathway (103). (B) Translation and transcription: The translation of viral ORF1a/b by host ribosomes produces two polyproteins, pp1a and pp1ab. proteolytic cleavage of pp1a and pp1ab by 2 cysteine proteases located within nsp3 (PLpro) and nsp5 (Mpro) releases 16 non- structural proteins, nsp1–11 (from pp1a) and nsp1–10, and nsp12–16 (from pp1ab), which are important for the viral replication and packaging of a new generation of viruses (103). Nsp12–16 form the replication-transcription complex (RTC). Nsp12 (RdRP), alongside its two cofactors nsp7 and nsp8, drives replication and transcription to produce full-length genomic and sub-genomic RNAs (sgRNAs). Nsp14 (Proofreading exonuclease) ensures error correction. Nsp13 (Helicase/NTPase and RNA 5′-triphosphatase activity), Nsp14 (N7-methyltransferase), and Nsp16+ Nsp10 (2′-O-methyltransferase complex) are required for Viral RNA capping to mimic host mRNA, increasing RNA stability and immune evasion (103). Ritonavir and Lopinavir (Antiretrovirals): decrease the viral load by inhibiting viral protease Mpro. Carmofur (1-hexylcarbamoyl-5-fluorouracil): an approved anticancer drug, have been shown to potently inhibit Mpro activity in vitro. (3) Remdesivir, Favipiravir, Galidesivir, and Levovir (Antivirals): halt the replication of the viral genome within the host cell by inhibiting the viral coded enzyme RNA-dependent RNA polymerase (RdRp). Ciclesonide (Glucocorticoid): It inhibits viral replication by interacting, directly or indirectly, with viral nsp15 (103). (C) Viral RNA and structural proteins (M, E, N, and S) are assembled into virions in host ER and Golgi apparatus, where M + E proteins form the viral envelope and N protein enhances RNA packaging. The mature viruses get released via exocytosis to infect other cells (103). CIM-834: binds and stabilizes the M protein in its short form in vitro, thereby inhibiting its oligomerization, which is crucial for successful viral particles assembly (103). ACE2 R, Angiotensin-converting enzyme 2 receptor; TMPRSS2, Transmembrane protease, serine 2; ORF, open frame reading; Nsp, non-structural proteins. Mpro, main protease; PLpro, papain-like protease; RdRP, RNA-dependent RNA polymerase; Rough ER, rough endoplasmic reticulum.

First, the viral S protein, primed by host transmembrane protease serine 2 (TMPRSS2), binds the ACE2 host cell receptor (46) (Figure 1). The S protein has two subunits: S1 and S2. The S1 subunit contains the ACE2-binding site, and the S2 subunit causes fusion between the viral envelope and the host cell membrane (46). The S1/S2 subunit boundary encodes a furin cleavage site, increasing the viral transmission rate and pathogenicity (46). Viral entry promotes cytokine release and contributes to cytokine storm (47). When infected with COVID-19, the host humoral response lasts ten or more days through IgA, IgM, and IgG antibodies (43). Detecting these three antibodies has demonstrated high accuracy in diagnosing COVID-19 (48).

Research shows a 79% homology between SARS-CoV and SARS-CoV-2 (49). Some studies indicated that the receptor binding domain (RBD) of SARS-CoV and −2 are identical. In contrast, other studies have demonstrated that SARS-CoV-2 has an affinity 5–20 times greater than SARS-CoV for the ACE2 receptor due to five amino acid residue substitutions that form additional hydrogen bonds and salt bridge interactions (43, 50). The higher ACE2 receptor concentrations lead to more efficient viral entry and higher replication rates (43). Lung and small intestine epithelial cells with high ACE2 expression are first infected (51, 52). Nineteen to twenty residues of ACE2, mainly in the N-terminal helix, are in contact with 17–19 residues of the SARS-CoV-2 RBD and interface through 13 hydrogen bonds and two salt bridges (53). Y505A is the most critical residue for SARS-CoV-2, as a single point mutation (Y505A) of the viral RBD was enough to eliminate any possible binding to the ACE2 receptor (53). SNPs in ACE2, such as the absence of ACE2 N546 in three out of 10,000 humans, alter expression and glycosylation states, which could raise the severity of symptoms (50, 54).

SARS-CoV-2 can enter the cell via endocytosis or by direct fusion with the plasma membrane, depending on the activation of the S-protein by the cellular TMPRSS2 protease (55). Upon entry into the host cell, viral RNA is released and subsequently replicated by biosynthesis, wherein maturation follows, and virions are released from the cell (56) (Figure 1). Generically, micro RNAs (miRNAs) are known to either increase or suppress viral RNA replication. Through RNA interference, miRNAs bind to complementary sequences of viral RNA and induce the formation of a silencing complex that inhibits the viral RNA and its protein expression. Long non-coding RNAs (lncRNAs) regulate gene expression in cytokine storms and can affect the expression of downstream targets by sponging miRNAs (56).

Immune response to SARS-CoV2 infection

The components of the host immune system seek to clear foreign microbes. While innate immune cells work broadly to eradicate invading particles, adaptive immune cells produce antibodies that recognize and deactivate extracellular antigens. In contrast, T-cells specifically target and kill infected cells or help other immune cells remove the infection. To eradicate the invading viruses, host innate immune cells secrete many chemical messengers, including proinflammatory cytokines and chemokines (57). The release of type I interferons (IFNs) among other proinflammatory cytokines is mediated through the activation of Pathogen Recognition Receptors (PRRs) by SARS-CoV-2 genetic material in a mechanism regulated by both Signal Transducer and Activator of Transcription 1 (STAT1) and Myeloid Differentiation Main Response Protein (MyD88) (58). Retinoic acid-inducible gene I-Like Receptors (RLRs) also sense the viral replicative intermediates and trigger IFN response that activates lung proinflammatory macrophages, which induce vascular leakage (58). The release of Viroporin-3a, an integral protein on the surface of SARS-CoV-2, activates nucleotide oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome pathway and IL-1β secretion from bone marrow-derived linking PAMP-PRR signaling in macrophages with cytokine release (59). On the other hand, damaged/dead host cells release their endogenous contents, which are recognized by PRRs, further aggravating the inflammatory response and creating a vicious cycle (60). Epithelial cells can fuel this inflammatory niche by producing other cytokines like MCP1/CCL2, CCL3, CXCL1, CXCL3, CXCL10, IL-8/CXCL8, IL-1β, and TNF-α, which draw macrophages and induce further tissue damage (61). These epithelial cells are spatially located in the vicinity of the HLA-DR-low/S100A-high macrophages, resulting in sustained contact between epithelial and immune cells (61). Eventually, tissue-quiescent macrophages are either polarized toward an inflammatory phenotype or replenished by infiltrating pro-inflammatory macrophages (62).

Uncertainty surrounds how T cell subsets contribute to SARS-CoV-2 pathogenesis and resolution. Mounting evidence suggests that the S protein is the chief antigenic protein that activates both humoral and cytotoxic T-cell responses. Xu and his colleagues showed that SARS-CoV-2-infected patients had significantly lower proportions of circulating CD4+ and CD8+ T lymphocytes, albeit being in a hyperactivation state. Moreover, higher percentages of cytotoxic CD8+T and T helper 17 (Th17) circulating cell subsets were linked to a severe inflammatory response (63). Along with the decline in peripheral T cell count, Diao et al. reported that SARS-CoV-2 also resulted in functional exhaustion of the remaining T cells. Given the imbalance between different T cell populations in many inflammatory disorders, more effort is needed to analyze the imbalance of the Th1/Th2 and Th17/regulatory T cell ratios in SARS-CoV-2 and disease progression (64). Specific IgA, IgM, and IgG responses comprise the host humoral defense mechanisms against the SARS-CoV-2 virus. Most COVID-19 patients develop a specific antibody (Ab) response 10 days or less after the onset of symptoms (65). Virologically neutralizing Abs are essential for the elimination of viruses. Tian et al. evaluated the newly developed 2019 coronavirus S protein (2019-nCoV) cross-reactivity against anti-SARS-CoV2 Abs (66). Collected from recently recovering patients, the study's authors demonstrated that the human CR3022 anti-SARS-CoV-2 monoclonal antibody bound to the 2019-nCoV RBD without overlapping with the ACE-2 binding site (66). They concluded that the CR3022 clone can be therapeutically used to tackle the current and emerging SARS-CoV-2 infections (66). In COVID-19-recovered patients, Ni and his group studied the cross-activation of SARS-CoV-2-specific humoral and cellular immunity (67). In this small cohort, neutralizing IgM and IgG against N protein and S-RBD had higher titer, which remained for at least 2 weeks. In vitro, assays revealed a shift toward high IFN-γ secreting N-specific T cells in SARS-CoV-2 convalescent subjects (67). Strikingly, SARS-CoV-2 neutralization Abs were statistically correlated with the number of N-specific T-cells, suggesting a dynamic association between humoral and cellular immunity during the battle against SARS-CoV-2 infection (67).

Hypercytokinemia and organ damage

In high-risk patients, SARS-CoV-2 infection may result in systemic and pulmonary inflammation and eventually MODS. The primary indicator of severe or critical SARS-CoV-2 pneumonia is organ failure (68). Patients with severe SARS-CoV-2 infection most frequently experience ARDS, shock, acute myocardial injury, liver injury, kidney injury, and MODS (68). It is commonly accepted that severe ARDS is the primary pathophysiology of critical COVID-19. The direct harm caused to target cells by SARS-CoV-2 and incorrect host responses, such as the immune-inflammatory response, may elicit multi-organ involvement (69). SARS and SARS-CoV-2 infections are characterized by fulminant MOD and high proinflammatory cytokine responses that may lead to hypercytokinemia, also known as cytokine storm, a phenomenon characterized by uncontrolled inflammation (70). Lung macrophages are believed to be the leading cause of hypercytokinemia by producing IL2, IL7, IL10, G-CSF, IP10, MCP1, MIP1A, and TNFα (71). In a retrospective study led by Chen and his colleagues, patients with moderate and severe SARS-CoV-2 infections had higher serum IL-2R, IL6, IL10, and TNFα cytokine profiles together with high risk of developing secondary hemophagocytic lymphohistiocytosis (72). Exaggerated and uncontrollable inflammation brought on by viral replication can harm the heart, liver, kidney, and central nervous system, resulting in sepsis, shock, or multiple organ failure (73). Higher numbers of tissue-infiltrating Th17 and cytotoxic T lymphocytes and acquired immune-induced proinflammatory responses may play a significant role in hypercytokinemia-induced tissue damage (71). Additionally, local production of proteolytic enzymes like elastase, collagenase, cathepsin, and matrix metalloproteinase may contribute to tissue and organ damage (73). In patients with SARS-CoV-2 infection and other inflammatory pathological processes, organ malfunction and tissue damage may result from the oxidative stress inflicted by the inflammatory macrophages and polymorphonuclear cells (74).

Zhou et al reported that, in COVID ICU patients, the aberrant pathogenic T helper 1 cells secreting higher levels of GM-CSF alongside the increased percentage of CD14+CD16+ inflammatory monocytes secreting a significantly higher levels of IL-6 could result in an inflammatory storm and severe lung damage, leading to lung functional disability, ARDS, and quick mortality (74). Xu et al. pointed out that SARS-COV-2 can lead to liver injury directly by infecting liver cells or indirectly by excessive inflammatory responses (74). Similarly, several mechanisms were reported to be involved in COVID-19-associated kidney injury, including the direct viral infection and the excessive inflammatory responses (74). Additionally, Mao et al. observed that markedly elevated proinflammatory cytokines in patients with severe COVID-19 may exert neurotoxic effects, contributing to neurological manifestations and skeletal muscle injury (74).

Immunosuppression

Lymphopenia, resulting from the increased lymphocyte infiltration into affected tissues, is a frequent symptom in COVID-19 patients and is associated with advanced disease stages and mortality. This state of immunosuppression can inhibit viral eradication and increase the likelihood of secondary infections, especially in SARS-CoV-2-hospitalized patients (5–15.5%) (75). A meta-analysis of 3,448 patients from 28 studies revealed that 14.3% of COVID-19 patients had subsequent secondary bacterial infections. These patients had higher SARS-CoV-2 viral loads, longer viral shedding, and reduced immune responses (76). This has a potential impact on cancer patients who are already or are going to receive anticancer drugs alone or in combination with immunosuppressives (77).

Pathophysiology of SARS-CoV-2

Respiratory system

There are three stages to the SARS-CoV-2 infection of the respiratory system. The first phase, which may be asymptomatic, affects cells lining the nasopharyngeal cavity without triggering a robust immune response (78). During the second stage, major airways are infected, and an early inflammatory reaction starts, whereas the virus invades type I and II pneumocytes in the third stage (78). While small cuboidal type II pneumocytes include “lamellar bodies” that secrete essential lung surfactants for normal alveolar functions, type I pneumocytes have a typical epithelial shape (79). Infection of type II pneumocytes upregulates antiviral genes, such as IFNs and specific ILs, and downregulates surfactant production genes (80). Escalated by high viral load, signaling in type II pneumocytes draws immune cells from the circulation and activates tissue-resident immune cells to activate pyroptosis in infected cells (78). This reaction worsens type I pneumocyte injury, causing the alveolar barrier to break down and cellular components to infiltrate the airways. The interstitium between the capillaries and the alveolar chamber expands as the immune response damages the alveolus and causes it to fill with a mixture of plasma exudate, dead cells, virus particles, inflammatory cells, and fibrin. Consequently, gas exchange is impaired, resulting in respiratory dysfunction that gives the disease, SARS, its name (78). Initially, it was believed that younger people are less likely to get infected because the lung tissue expression of ACE-2 increases with age. However, the data appears inconclusive due to the lack of age-dependent variation in the receptor expression (81, 82).

Cardiovascular system

Thrombotic events, including disseminated intravascular coagulation and venous thromboembolism, are the primary comorbidities in COVID-19 patients (83). This is because SARS-CoV-2 infection induces severe hematological changes like leukopenia (80%), thrombocytopenia (30%), and anemia (12%) (84) together with a rise in inflammatory cytokines including IL-6, IL-2, IL-7, interferon-ɤ, and TNFα, and high plasma levels of the thrombotic risk markers D-dimer, procalcitonin, and C-reactive protein (84). These biochemical alterations induce chronic hypertension that, in combination with the prothrombotic state, dramatically raises the risk of thromboembolism (85). Co-factors, such as cardiovascular diseases (coronary heart disease) or chronic diseases, such as type-2 Diabetes Mellitus (T2DM), are correlated with poor outcomes (84). Importantly, autopsy sequencing proved that SARS-CoV-2 infection is not a direct cause of the heart failure observed in terminally sick patients (85). Instead, the SARS-CoV-2-induced systemic inflammation seems to be the primary cause of cardiac dysfunction, with tissue endothelia's key role in disease pathophysiology (84).

Gastrointestinal tract

Dental complications

Originating from sputum or nasal cavities, SARS-CoV-2-infected individuals with typical symptoms had detectable viral loads in their saliva (86). Sequencing of tissue biopsies confirmed the positivity of the SARS-CoV-2 receptor in all oral cavity cell types and salivary gland epithelia of patients with COVID-19 infection, suggesting that the mouth may serve as both a point of infection and a virus reservoir (87) and explaining specific COVID-19 related symptoms like dysgeusia and ageusia (88). Over months, numerous lesions, including ulcers, erythema, and spontaneous bleeding, have been reported in the oral cavity of infected patients. Risk factors for dental complications included implants, xerostomia, poor oral hygiene, opportunistic infections, stress, vascular impairment, and COVID-19-related hyperinflammatory response (88).

Hepatic complications

Impact on disease course

According to Chan SL & Kudo M, different reports of COVID-19 stated that 2–11% of COVID-19 patients had comorbid liver diseases (89). In patients with compromised hepatic reserves, SARS-CoV-2 may cause additional hepatic injury or hepatic decompensation. Additionally, SARS-CoV2's potential immunosuppressive property might lead to viral reactivation in COVID-19 patients with chronic viral hepatitis (89) (Figure 1). Antiretrovirals such as lopinavir/ritonavir, antifungal agents, antibiotics, and other drugs required to treat SARS-CoV-2 and its associated inflammation may cause drug-induced liver injury or reactivate hepatitis B virus (89, 90). This drug-induced cytotoxic effect, alongside the suggested SARS-CoV-2 viral replication in hepatocytes, can damage the remaining parenchyma in patients with liver cirrhosis and worsen their prognosis (90). The COVID-19 lockdown caused social isolation, sedentary behavior, and an increase in processed food consumption, all of which are unfavorable metabolic antagonists in non-alcoholic fatty liver disease (NAFLD) patients (91).

Impact on patient care

During the physical distancing at the beginning of the COVID-19 pandemic, resources were diverted for the sickest patients to decrease hospital utilization. Stable patients' routine care was put off, lessening the spread of SARS-CoV-2 (92). Hepatocellular carcinoma (HCC) and varices screening were converted from all-comers to high-risk patients only. As a result, these delays increased the risk of variceal hemorrhage and, in approximately 25% of patients with biologically aggressive disease, raised the likelihood of HCC being discovered at a later stage (92). They also anticipated that “returning to normal” after physical distancing would be difficult with more decompensations and morbidity and care systems being overburdened by the backlog of put-off care. Patients who were incorrectly classified as low risk in the first wave would start to present with decompensations, requiring more resources and raising the possibility of cascade deferrals for the presently asymptomatic patients. Complications of delayed screening procedures, failure to diagnose HCC at earlier stages, and medical therapy complications due to lack of timely lab work all would lead to the transformation of the curable into the incurable (92). In an Italian study that included 42 HCC patients, it was reported that the pandemic caused a treatment delay of 2 months or longer in 11 patients (26%) & change in treatment plan from surgical resection to thermal ablations in 3 patients. It is unknown whether or not these delays and modifications will affect the long-term outcomes for these patients (93). Several factors contributed to a dramatic decrease in the number of liver transplantation procedures during the COVID-19 pandemic, including the cancellation of elective therapeutic procedures such as living donor liver transplantations, uncertainty regarding the feasibility of intensive care beds, blood products or ventilators required to accept deceased donor's organ for the waitlisted patients, uncertainty regarding the test-accuracy for SARS-CoV-2 and its transmission, conversion of several transplantation centers into COVID-19 units, and the quarantine travel restrictions leading to increase in waitlist mortality (92). Viral hepatitis was no exception in terms of the redirected attention and resources from other diseases to COVID-19. Since most elimination programs for viral hepatitis were put on hold or stopped altogether, the WHO's goal of eliminating HBV and HCV and decreasing their mortality by 2030 has been hindered. One study proposed that a 1-year delay in diagnosis and treatment of viral hepatitis will globally result in 72,300 deaths from HCV and an additional 44,800 HCC cases in the next 10 years (91).

Lower GIT complications

Approximately 20% of SARS-CoV-2-infected patients exhibit gastrointestinal symptoms such as nausea, vomiting, and diarrhea; this number jumps to 50% in hospitalized patients. It is now thought that SARS-CoV-2 directly infects enterocytes, with the intestinal epithelium having the highest ACE-2 expression level (94). This might explain why seriously diseased patients witness symptoms resembling those of inflammatory bowel diseases like Crohn's disease and irritable bowel syndrome and justify the presence of the viral genetic material in the feces of infected patients even if they didn't develop typical SARS-CoV-2 symptoms (94). Moreover, changes in the immune microenvironment within gastrointestinal (GI) tract organs and their microbiota resemble the proinflammatory characteristics of SARS-CoV-2 -infected lungs (94, 95).

Central nervous system

Approximately 30% of infected individuals experience neurological symptoms, which can range from minor issues like headaches to serious ones like cerebrovascular infarction. Three potential causes of COVID-9's pathophysiology manifestation in the neurological system include chronic inflammation, underlying coagulopathy-induced thrombotic events, and direct infection of nervous system cells. There is evidence that SARS-CoV-2 crosses the blood-brain barrier since it has been detected in the patient's brain tissue, and upregulation of interferon signaling pathways of the neurovascular compartments has been shown (96). Furthermore, brain capillary endothelial-like cells were infected in in vitro models. Besides severe brain tissue infections, taste and smell loss are typically categorized as neurological complaints.

Dermatologic manifestations

The COVID-19 cutaneous symptoms are primarily divided into six categories of injury: confluent erythematous/maculopapular/morbilliform rash, urticarial rash, papulovesicular exanthem, chilblain-like acral pattern, livedo reticularis/racemose-like pattern, and purpuric pattern (97). These injury patterns, which frequently include endothelial damage and perivascular inflammation of dermal arteries, are likely the result of cutaneous reactions to circulating viral antigens. Erythematous or maculopapular eruptions appear after the onset of systemic symptoms in nearly 50% of patients with SARS-CoV-2-related rashes. These lesions typically have pruritic symptoms, symmetrical distribution over the trunk, and centrifugal propagation. Despite the paucity of data, these eruptions appear to be caused by vascular damage and frequently have perivascular lymphocytic or neutrophilic infiltrates (97). The trunk and limbs are where urticarial rashes are most noticeable, and they may also be accompanied by angioedema or urticarial vasculitis (97). They occasionally appear before pulmonary illness presentations in individuals, although they typically appear simultaneously as other systemic symptoms. Papulovesicular exanthems frequently have vesicles dispersed around the trunk, either with or without pruritus. Biopsies show endothelial inflammation in dermal arteries and epidermal acantholysis with inflated keratinocytes (98). Endothelial damage and thromboses are frequently present in eruptions with the acral pattern, livedo reticularis, and racemose-like patterns. These eruptions may be brought on by IFN-mediated inflammatory responses or endothelial injury related to circulating viral particles (98). Chilblain-like lesions manifest as violaceous plaques that primarily affect the feet but can also occasionally involve the hands or ears. Unlike other COVID-19 dermatologic manifestations, these lesions are typically unpleasant or itchy and tend to affect people with little or no systemic symptoms (98). The livedo reticularis/racemose-like pattern is characterized by lace-like patches with a dusky blue discoloration and most likely signifies sluggish blood flow brought on by thrombosis of small superficial veins in the dermis (98).

Pregnancy

It is crucial to assess the possibility of severe consequences from SARS-CoV-2 infection in pregnant women and fetuses/newborns. Clinical traits of pregnant COVID-19 carriers have been examined in several original investigations and systematic reviews (99). Notably, most studies compare pregnant women with COVID-19 to non-pregnant women of reproductive age in terms of clinical features and death rates. In contrast, SARS-CoV and other respiratory viruses have shown different symptoms (100). Some have hypothesized that this is due to the natural immunological adjustments during pregnancy, preventing COVID-19 from escalating to the hyperinflammatory phase. Although there is evidence of moderate COVID-19 in pregnant patients, a new analysis by the CDC implies that pregnant women may be at a higher risk for more severe consequences, suggesting that a higher percentage of pregnant women with COVID-19 require hospitalization compared to non-pregnant women (101). However, the lack of information indicating whether hospital admission was brought on by COVID-19 sickness or symptoms related to pregnancy limits this study and makes interpretation more difficult (101). In addition to these studies, there is mounting evidence that pregnant SARS-CoV-2-infected women experience enhanced rates of miscarriage and hypertension, indicating placental involvement (100). Most research has found no evidence that the placenta contains SARS-CoV-2 RNA. However, a recent case report revealed SARS-CoV-2 in the syncytiotrophoblast cells of a COVID-19 patient who was pregnant and had preeclampsia in the second trimester of the pregnancy. It also indicates that there is a distinct association between the laboratory profile seen in preeclamptic pregnant individuals and COVID-19, raising issues about common disease mechanisms (100). The latest research suggests that vertical transmission of SARS-CoV-2 is improbable, which is an important final point to make. However, as previously mentioned, SARS-CoV-2 can damage the placenta dramatically and consequently adversely affect fetal development. To more fully understand the clinical profile of COVID-19 during each trimester of pregnancy, additional study is required (100).

Pediatrics

In a case study of more than 2,000 children with suspected or confirmed COVID-19, only 0.6% of the symptomatic individuals proceeded to ARDS or Multiple Organ Failure (MOF), while 5% exhibited dyspnea or hypoxemia (102). Interestingly, the laboratory profile of pediatric COVID-19 is distinctive from that of adults in cases with mild infections. However, a meta-analysis of 24 studies, including 624 pediatric cases with PCR-proven COVID-19, has shown common laboratory abnormalities in moderate and severe disease courses regarding CRP, procalcitonin, and LDH but not in lymphocyte count (102). One explanation of such discrepancy might be the disparate standards employed to interpret laboratory testing in pediatrics (102). Moreover, a recent study of a small cohort of previously healthy children and teenagers who developed an inflammatory profile linked to COVID-19 described a special cytokine pattern characterized by elevated IL-6 and IL-10 production, as well as increased IFN signaling components with no increase in TNF-α which is common among adults (103). Pediatric COVID-19 was also associated with GI symptoms and cutaneous signs (104). In response to the unmet need to accurately identify pediatric COVID-19, WHO and other regulatory authorities have created a preliminary case description known as Multisystem Inflammatory Disorder in Children and Adolescents (MIS-C) (103). Further studies are necessary to determine the applied protective measures against pediatric COVID-19 and the underlying causative mechanisms behind MIS-C in children.

Lines of treatment for COVID-19

SARS-CoV-2 lifecycle and therapeutic targets

For most people, COVID-19 can be treated like a common cold: rest, fluid intake, and over-the-counter medicine to reduce symptoms like fever and congestion. For those who experience more severe symptoms that require hospitalization, respiratory support is the primary focus of treatment with therapeutics. Multiple clinical trials were performed to test the efficacy of different treatment options, including antiviral therapies, corticosteroids, immunotherapies, dietary supplements, and more.

Antiviral therapeutics

Antiviral therapeutics were investigated to treat COVID-19 as they function in a variety of mechanisms (105). Depending on the disease severity, infection can be divided into three stages: non-severe, severe, and critical. In the non-severe stage, viral replication occurs enough that antiviral therapeutics can treat the infection (106). These antiviral agents can be categorized according to their mechanism of action: those that inhibit viral entry into host cells, those that block viral replication, and those that modulate the immune response. Among the drugs that specifically target viral replication are Paxlovid, remdesivir, molnupiravir, and ribavirin (106).

Remdesivir, in particular, has demonstrated clinical benefits (Figure 1). When used in combination with supportive care, it has been associated with shorter hospital stays and a significantly reduced mortality rate (105). In a study involving non-hospitalized patients who were at high risk for developing severe disease, a 3-day course of remdesivir led to an 87% reduction in the risk of COVID-19-related hospitalization or death from any cause by day 28, compared to those who received a placebo (106). According to WHO guidelines from July 14, 2022, antiviral drugs such as nirmatrelvir-ritonavir (Paxlovid®) are strongly recommended. Polymerase inhibitors, such as molnupiravir and remdesivir, are weak or conditional recommendations (107). Monoclonal antibody (mAb) therapies that target viral entry into host cells include casirivimab + imdevimab, bamlanivimab, sotrovimab, and bebtelovimab. These agents have been used both for treatment and prophylaxis of COVID-19. Their effectiveness can vary depending on circulating variants, and resistance can emerge, especially in immunocompromised individuals. Therefore, virological monitoring in these patients is crucial. In such cases, the use of combination monoclonal antibody therapies is highly recommended to enhance efficacy and reduce the risk of resistance development (107).

Corticosteroids

The use of corticosteroids in treating COVID-19 has been the main question since the beginning of the pandemic. COVID-19 infection occasionally develops aggressive inflammatory responses that progress to life-threatening respiratory problems, including severe pneumonia, cytokine storm, and ARDS. It has been reported that patients in severe and critical stages have higher serum cytokine amounts (105, 108). Corticosteroids act as immunosuppressive agents for excessive inflammatory responses and prevent the progression of hyperinflammation. Once infection occurs, the virus infiltrates host cells through interactions with angiotensin-converting enzyme-2 (ACE-2) receptors, initiating an immune response. The release of cytokines in large quantities results in a cytokine storm. Cytokines such as interleukins (IL-1, IL-2, IL-6, IL-8, dan IL-12) and interferons (INF-α, INF-β, dan INF-γ) play a significant role in recruiting immune cells, including macrophages and lymphocytes (108, 109).

Corticosteroids suppress the activation and recruitment of these immune cells through their immunosuppressive effects, thereby preventing the deleterious effects of cytokine storms that can lead to dyspnea, multi-organ failure, and death (109).

Therefore, anti-inflammatory drugs such as dexamethasone, hydrocortisone, or prednisolone are essential for cytokine storm suppression (110). According to WHO guidelines on 13 January 2023, corticosteroids are recommended only in hospitalized patients (111). The data does not support usage in non-hospitalized patients (109).

Immunomodulatory therapeutics

Regulation of immune responses against SARS-CoV-2 was one of the most important aspects of disease pathogenesis, particularly in severe and critical stages (112).

Interleukin-inhibitor: tocilizumab

After infection, cytokine release syndrome (CRS), with excessive immune responses and subsequent release of pro-inflammatory mediators, chemokines, and cytokines, is observed. This plays a crucial role in the severity of COVID-19. IL-6 plasma level was high, especially in severe cases. Tocilizumab is an anti-human IL-6 receptor monoclonal antibody which antagonizing the IL-6 receptor, both IL-6 soluble receptor (sIL-6R) and membrane receptor (mIL-6R) (113). It can bind the membrane-bound IL-6 receptor and inhibit signal transduction (114). Patients who developed COVID-19-related CRS could use tocilizumab with promising suppressive action (114). A cohort study discussed the pathological role for the interleukin-6 (IL-6) pathway in mediating structural and functional delirium-like phenotypes. They tested if tocilizumab is associated with a reduction in delirium/coma prevalence in critically ill patients with COVID-19. Delirium was assessed using the Confusion Assessment Method for the Intensive Care Unit (CAM-ICU), with a positive score indicating delirium. Tocilizumab was associated with a significantly greater number of days alive without delirium/coma (113).

Janus kinase inhibitor: baricitinib

The pathophysiology of COVID-19 involves a signaling pathway based on the Janus kinases (JAKs) and activator of transcription (STAT) pathways (115). Janus kinase inhibitors (JAKi) have been approved for various immune-mediated inflammatory diseases (115). Baricitinib is a selective JAKi inhibitor with a proven anti-inflammatory effect (303). An international, multicenter, double-blind, randomized, placebo-controlled trial evaluated the benefits and safety of baricitinib with standard of care (corticosteroids and remdesivir) for hospitalized patients. Thus, it has been suggested that baricitinib could be a good option to decrease mortality when used with standard of care (116).

A retrospective study suggested survival and safety are significantly better for baricitinib compared to tocilizumab in severe COVID-19. This study included 400 hospitalized patients with severe COVID-19 (116).

Chloroquine and hydroxychloroquine

Chloroquine and Hydroxychloroquine had an extensive record of malaria treatment and its prevention (117) (Figure 1). Although they have been recommended in COVID-19 treatment based on experiments in Vero E6 and A549-ACE2+ cells (118121), it has been shown that chloroquine is inactive in the relevant lung adenocarcinoma Calu-3 cell line (122, 123). Furthermore, many clinical trials have failed to prove any positive effects of these drugs on clinical status or lower mortality (124). When hospitalized patients with respiratory illness were treated with hydroxychloroquine, compared with a placebo, there was no clinical significance in improving clinical status. Therefore, these results do not support the use of these drugs in COVID-19 treatment (125). WHO guidelines on March 24, 2023, strongly recommended against administering hydroxychloroquine to prevent COVID-19-related outcomes (126).

Anti-coagulants

COVID-19 leads to high levels of mortality because it damages some vital organs, including the cardiovascular and coagulation systems (127). In severe and/or critical stages, patients with pneumonia are highly prone to thrombotic complications (128). Anticoagulants, especially low molecular weight heparin, are associated with better prognosis in severe COVID-19 patients and decreased mortality risk (129).

Dietary supplements

Various vitamins and minerals, such as vitamin C and zinc, support patients' immune status. Vitamin D deficiency might increase susceptibility to diseases (130). Vitamin C is an antioxidant that supports the immune system and protects against infections, which may render it an effective treatment addition (131). Zinc is a trace mineral that has a role in response to viral infection. Zinc deficiency affects immune function by impairing the formation and maturation of lymphocytes (132). A study showed that people with zinc deficiency have a higher risk of respiratory diseases (133). Vitamin D is known for its role in protecting bones, and it supports the development of some immune cells (131). A randomized clinical trial found that a high dose of vitamin D decreased CD4+ T-cell activation, worsening viral infections (134).

Natural and synthetic compounds in the treatment of COVID-19

Since the emergence of the pandemic, scientific efforts have been devoted to working out the best treatment regimen for COVID-19. Computational analysis alongside assays on human cells and in mammalian experimental models generated an immense repository of useful information and presented novel antiviral strategies. Mining the data for COVID-19-tested drugs in 199 peer-reviewed articles published between 2020 through 2023 identified 231 natural and 169 synthetic compounds that played protective and/or therapeutic roles against COVID-19. Among these compounds, 292 were tested using in silico software, 50 were validated in vitro, 58 were tested both in vivo and in vitro, and 22 underwent clinical trials. In the following section, we sought to compile some details regarding these compounds as in Supplementary Table S1. Natural products have proven efficiency against COVID-19. Several natural products from different sources (plants, microbes, and marine) were effective against COVID-19. Natural products have been either tested as extracts or in the pure form of the compounds (135153, 168, 170).

In silico analysis

Currently, artificial intelligence is being used in the drug discovery of natural products. It has been applied to predict the macromolecular targets of natural products, the structural characterization of natural products, and the selection of natural products as drug candidates. Computational drug discovery approaches such as docking, clustering, bioactivity fingerprints, pharmacophores, and machine learning shed light on the possible targets for natural products. Moreover, the potential binding modes and the affinity of natural products could be studied via molecular docking and molecular dynamics (154). Molecular docking software, such as Auto Dock Vina, Gold, Glide, MOE, and AutoDock software, provides indispensable information about the binding of small molecules to a specific protein macromolecule in its biologically active 3D form. Given the advances of artificial intelligence and machine learning, bioinformaticians and biostatisticians were able to analyze the interaction between natural and synthetic compounds with SARS-CoV-2 key proteins.

Mpro target protein

Auto Dock Vina software has identified Hypericin (155), Isohypericin (155), and Anthrachinolinchinon (156) as top hits against Mpro protein. Docking studies run by PyRx software showed that Bemcentinib (157) effectively inhibits Mpro. While Gold software ranked Rutin (158) and Epirubicin (158) with the highest docking scores, Glide score software flagged compounds Pectolinarin (159) and Quercetagetin (160) are potential therapeutic candidates against Mpro. Indomethacin (161) and Bislatumlide A (162) had the top docking scores using AutoDock software. MOE software suggested Methyl rosmarinate (163) as the top hit. Finally, Acetoside and Luteolin 7-rutinoside (164) had highest docking scores using iGEMDOCK, while 2,3-Dihydroamentoflavone (165) was highlighted by MTi AutoDock software.

ACE2 protein

Hesperidin was the best hit using Auto Dock Vina, while Kobophenol A and Andrographolide ranked first using AutoDock (304). Furthermore, 5-O-Feruloyl-quinic acid had the highest docking score against ACE2 protein using PyRx, while Isothymol strongly bound to ACE2 protein, as suggested by MOE (305).

Nsp15 protein

AutoDock software identified Indomethacin (161) and Demethoxycurcumin (166) as the compounds with the highest docking scores, while Catechin-7-o-gallate (167) had the top score as calculated by PyRx. Analyzing different compounds against Nsp15 protein using AutoDock Vina detected Cnicin (168) and Glyasperin A (169) to have the highest scores.

Nsp16 protein

Compounds that showed the highest docking scores in the in silico studies were: Daphnorin (170) and Glycycoumarin (170), using Autodock Vina; Amentoflavone (171) and Baicalin (171) using PyRx; Indomethacin (161) using AutoDock.

TMRSST protein

The best hits are Myricetin (172), Meloxicam (172), and Columbin (172) using Autodock Vina; Withanoside-IV (173), Withanoside V (173) using LeDock (174).

NSP10-NSP16

The best hits are Withanolide (175) and Dolutegravir (175) using Autodock Vina and Chlorogenin (167) using PyRx.

PLpro protein

The compounds with highest docking scores were: Schaftoside (176) and 1-Hydroxyaleuritolic acid 3-p-hydroxybenzoate (177) using AutoDock Vina; UKR1129266 (178) using C-docker; Cichoriin (179) using COVID-19 Docking Server; Theaflavin 3-gallate (TF2a) (180) using smina server; and Indomethacin (161) using AutoDock.

RdRp target

Amentoflavone (167), Emblicanin A (181), and Cyanin (167), were the top 3 hits using PyRx; Taiwanhomoflavone A (182), Cnicin (168), and Nympholide A (182) were identified using AutoDock Vina; Indomethacin (161) was detected by AutoDock; Theaflavin 3,3′ digallate(TF3) (180) using smina server; Cichoriin (179) using COVID-19 Docking Server; and Tetrahydroxycurcumin (183), Andrographidine C (184), and Silibinin (185) using Glide.

S-protein

Glycyrrhizin (169) was the best hit using AutoDock Vina. Agathisflavone (167), and Catechin-7-o-gallate (167) had the top docking scores using PyRx. Indomethacin (161), Speciophylline (186), and Uncarine F (186) were highlighted using AutoDock. Silibinin (185) was chosen using Glide. Finally, Argus lab and CLUSPRO 2.0. identified 1-(2,3- dihydrobenzo[b](1, 4)dioxin-6-yl)-2-(furan-2-yl)-4,5-diphenyl-1 H-imidazole (DDFDI) (187) and Urtica dioica agglutinin (188) as top hits, respectively.

In vitro studies

Alongside computer-based analyses, many research groups focused on analyzing the effect of different natural and synthetic compounds using wet laboratory settings with the hope of discovering or repurposing an existing compound to treat patients with COVID-19.

Mpro target protein

For example, (1R,2S,5S)-6,6-dimethyl-N-((S)-1-oxo-3-((S)-2-oxopyrrolidin-3-yl)propan-2-yl)-3-(2-(4- (trifluoromethoxy) phenoxy) acetyl)-3-azabicyclo[3.1.0]hexane-2-carboxamide (189), Silibinin (185), and GC376 (7) (190) compounds had IC50 of 0.015 μM, 0.021 μM, and 0.15 μM against Mpro target protein, respectively. Compounds GC376 (190), (1R,2S,5S)-6,6-dimethyl-N-((S)-1-oxo-3-((S)-2-oxopyrrolidin-3-yl)propan-2-yl)-3-(2-(4-(trifluoromethoxy)phenoxy)acetyl)-3-azabicyclo [3.1.0] hexane-2-carboxamide, and (1S,3aR,6aS)-2-(2-(2,4-dichlorophenoxy)acetyl)-N-((S)-1-oxo-3-((S)-2-oxopyrrolidin-3-yl)propan-2-yl)octahydrocyclopenta[c]pyrrole-1-carboxamide (189) had the lowest IC50 against SARS-CoV-2 targeting Mpro with IC50 of 0.70 μM, 1.2 nM, and 1.1 nM, respectively.

ACE-2 protein

Compounds effective against ACE-2 protein included Licarin B and Licarin A with IC50 of 430.11 nM and 3.59 μM, respectively (191).

S-protein

Pomegranate peel extract (PPE) (192), Kobophenol A (193), and Silibinin (185) have the lowest IC50 against S-protein with IC50 of 0.049 mg/ml, 1.81 μM, 0.029 μM, respectively. Cysteamine HCl (194) had the lowest IC50 against SARS-CoV-2 targeting S-protein.

PLpro target

Dihydrotanshinone I (195), Sepantronium bromide (196), Schaftoside (176), and UKR1129266 (178) had inhibitory activity against PLpro with IC50 of 0.59 μM, 2.47 μmol/L, 3.91 μmol/L, and 0.90 μM, respectively.

RdRp target

Silibinin (185) showed the lowest inhibitory activity against RdRp with IC50 of 0.042 μM.

TMRSST protein

Withanoside-IV showed in vitro activity against SARS-CoV-2 targeting TMRSST with %inhibition of 45.03% and 44.79% for E (envelope) and N (nucleocapsid), respectively (173).

In vivo studies

Based on the successful role against COVID-19 in vitro, the antiviral effect of some natural and synthetic compounds was tested in experimental mammalian models.

Mpro target protein

Indeed, 6-Amino-1-(4-chlorophenyl)-4-(2,4-dichlorophenyl)-3-methyl-1,4- dihydropyrano[2,3-c]pyrazole-5-carbonitrile (197), FB2001 (198), Nirmatrelvir (199, 200), and Ensitrelvir (201) inhibited SARS-CoV-2 in vivo studies.

ACE-2 protein

Dalbavancin had in vivo activity in rhesus macaque models with P-value of 0.0042 (202). PLpro protein

Aurintricarboxylic acid (203) had a good inhibitory activity for PLpro protein in vivo studies.

RdRptarget

(3R,4R,5R)-2-(4-aminopyrrolo[2,1-f][1,2,4]triazin-7-yl)-2-cyano- 5-((isobutyryloxy)methyl)tetrahydrofuran-3,4-diylbis (2-methylpropanoate) (204) and Molnupiravir (205, 206) had in vivo activity against SARS-CoV2.

S-protein

H84T-BanLec (207) showed inhibitory activity against SARS-CoV-2 in vivo studies using a golden Syrian hamster model with a P-value of < 0.05 targeting S-protein.

Clinical trials

Vaccines

In January 2020, Chinese scientists completed and submitted the first SARS-CoV-2 genome sequence to the NIH's GenBank database (208). Prior to the COVID-19 pandemic, the fastest vaccine development on record was for mumps in the mid-20th century, which took four years (209). Remarkably, less than a year after the SARS-CoV-2 sequence was published, Pfizer-BioNTech released the first COVID-19 vaccine for individuals aged 16 and older in December 2020 (210). This unprecedented speed was made possible by decades of prior research, substantial funding, and expedited regulatory pathways (211). Currently, five major platforms dominate COVID-19 vaccine development: mRNA, non-replicating adenovirus vectors, inactivated virus, protein-based, and DNA vaccines (212, 213) (Table 1).

Among these, mRNA vaccines are categorized into three types: circular RNA (circRNA), non-replicating mRNA (NRM), and self-amplifying mRNA (SAM) (214). These vaccines deliver mRNA into the cytosol of host cells, where it serves as a template for antigen production, triggering an immune response (215). Despite being studied for over two decades, mRNA technology faced challenges due to ribonuclease activity and mRNA instability. These issues were overcome with the development of lipid nanoparticle (LNP) delivery systems and chemical modifications to the mRNA itself (214, 216). mRNA vaccines do not require a live virus and can be designed within days of sequencing a viral genome (217). The use of LNPs has further enhanced the stability and delivery of mRNA vaccines, and their non-integrating nature eliminates the risk of infection (218).

Circular RNA (circRNA) is a covalently closed eukaryotic nucleoside ring that provides protection from degradation via endonucleases because of a lack of termini, yielding a more stable molecule than a linear RNA molecule (219, 220). CircRNA does not possess 5′ or 3′ ends, and naturally occurring circRNA can be protein-coding as well as non-coding (220). CircRNA is generated through back-splicing of pre-mRNA and, until recently, was viewed as a splicing error by-product (221). Current circRNA platforms circumvent cap-dependent translation of RNA by designing templates with either an upstream m6A modification or by introducing an internal ribosome entry site (219). Current research groups have successfully engineered circRNA COVID-19 vaccines that demonstrate prolonged antigen translation due to enhanced stability, as compared to linear mRNA vaccines (222). Interestingly, naturally occurring circRNA has been shown to alter gene expression through the regulation of nuclear transcription, RNA-binding proteins, and microRNA (223).

Both non-replicating mRNA (NRM) and self-amplifying mRNA (SAM) vaccines are linear and possess a 5′ 7-methylguanosine cap, 3′ poly(A) tail, a coding sequence inside an open reading frame, and a 5′ and 3′ untranslated region (224, 225). The 5′ cap both serves as a binding site for eukaryotic translation initiation complex cap-binding protein (eIF4E) and aids in protecting the mRNA transcript from nuclease-mediated hydrolysis (225). The poly(A) tail shields the transcript from degradation while also regulating translational efficiency based on the tail's length (215).

However, SAMs have a large open reading frame with desired antigen genes replacing structural genes from the original viral genome, which eliminates the risk of viral infection while also contributing to SAMs overall larger size when compared to NRM (226). Additionally, SAM vaccines possess viral non-structural protein genes, which encode for the replication machinery capable of producing the RNA transcript (227). Comparatively, NRM vaccines may require higher treatment doses when compared to SAM vaccines because the amount of antigen manufactured is directly dependent on the available mRNA transcripts provided by the treatment. Conversely, SAM vaccines can duplicate mRNA transcripts, leading to higher antigen expression within treated cells while also enhancing the innate immune response by propagating adjuvants (227, 228). However, NRM vaccine's advantage over its SAM counterpart stems from its shorter and more simplistic transcript, which most COVID-19 vaccines available are based on (214).

Adenoviruses (AVs) can elicit a cellular immune response, humoral immune response, or a combination of both when infecting host cells (229) (Table 1). AVs contain double-stranded DNA within an icosahedral capsid lacking an envelope (230). AV vaccines are capable of stimulating PRRs, such as toll-like receptors, which promote an innate immune response without the need for adjuvants. However, the inflammatory response is not extreme enough to cause a cytokine storm (231). Additionally, AV vaccines are incapable of replication because the E1 and E3 gene cassettes—which are imperative for viral replication—are replaced with the desired antigen sequence (232). One current COVID-19 AV vaccine is based on the type 5 vector and is designed to be inhaled, mimicking the transmission of COVID-19 into humans (233).

Inactivated virus (IV) vaccines are a traditional approach to generating an immune response that has been used in milestone treatments, such as Jonas Salk's inactivated polio vaccine (234). The immunogen for IV vaccines is the whole virus, which is inactivated with heat, chemicals, or radiation (235, 236); this confers an advantage when considering humoral responses because an abundance of epitopes from the whole virus can in turn generate many unique antibodies when compared to an mRNA vaccine that only causes expression of spike proteins (235) (Table 1).

Protein subunit vaccines utilize viral products to trigger an immune response within a host (237) (Table 1).

COVID-19 subunit vaccines typically target antigenic parts of the virus-like the RBD—or the full-length spike proteins—like S1 (238). The inclusion of only a viral product—not the entire virus—can reduce the chance of an unintended autoimmune or inflammatory reaction (237). Additionally, adjuvants can be conjugated with epitopes in a subunit vaccine to enhance the immune response (239) (Table 1).

DNA vaccines are like the previously discussed RNA vaccines. However, the nucleic acid in DNA vaccines encoding viral antigens is translocated into the nucleus for transcription after entering the host cell (216, 240). This poses a possible risk of integration into the host genome compared to RNA vaccines, which typically require freezer storage (306). DNA vaccines have improved stability and maybe a better candidate for treatment in resource-limited areas (240242). DNA vaccines can also activate both humoral and cellular immune responses (243) (Table 1).

As of March 30, 2023, the type, number, and relative percent of the 183 candidate vaccines in the clinical phase include protein subunit (59, 32%), viral non-replicating vector (25, 14%), DNA (17, 9%), inactivated virus (22, 12%), RNA (43, 24%), and others (244). Additionally, there are 199 vaccines in pre-clinical development (244). There are currently two types of vaccines approved by the Food and Drug Administration (FDA): mRNA and protein subunit vaccine. Moderna and Pfizer-BioNTech produce the mRNA vaccine, while Novavax manufactures the protein subunit vaccine. However, J&J/Janssen produced an FDA-approved viral vector vaccine, which expired in May 2023 (245). The efficacy of Pfizer-BioNTech's vaccine is 95%, Moderna's is 94.1%, and J&J/Janssen's was 83.5% (246).

COVID-19 vaccines are diligently constructed to meet the FDA's standards; however, side effects have been reported (247). Common side effects are influenced by age, but for adults, they may include: swelling and pain at the injection site, chills, fever, tiredness, etc. (248). Albeit rare, adverse events may occur following vaccination. This includes anaphylaxis, thrombosis, myocarditis, pericarditis, and Guillain-Barré Syndrome (249).

Artificial intelligence

The advancement of data science has significantly contributed to a breakthrough led by artificial intelligence (AI) applications such as machine learning (ML) or deep learning (DL) in various aspects, especially in the medical and pharmaceutical fields. In this context, with the viral infection outbreak causing COVID-19 in 2019, this pandemic has posed a multi-directional challenge, whether in medicine, defense, information technology, or even politics and ideologies. The COVID-19 pandemic has dramatically accelerated the adoption of AI applications in emergencies, particularly in tracing epidemiological peaks or operating within complex scenarios involving healthcare (250). The pandemic has demonstrated that the diversity of AI-driven tools' potential can eradicate health disparities on the one hand and upgrade the efficiency of health systems on the other. Studies have shown that AI interventions during the pandemic outbreak have improved various pathways, including identifying and diagnosing cases using trained deep learning algorithms, which provide results around the clock, about 135 times faster than a radiologist (251). Using neural networks and ML models also contributed to implementing preventive measures such as social distancing, identifying people without a protective face mask through their eye line, and alerting citizens to avoid crowded public places or to wear face masks (252). Furthermore, AI applications have delivered new tools to track the spread of the virus, particularly in hotspot areas. For example, in Japan, scientists developed a disease spread simulation model and GPS data miner to analyze the hotspot detection of COVID-19 infection using mobile phone location data (253). Also, a study reported that ML models have been relied upon to carry out the tasks of early identification and diagnosis of the infection based on clinical information, not CT images. One of these ML tools has been published online (https://intensivecare.shinyapps.io/COVID19/) (254). In the same context, several studies have shown the promising use of DL and ML models to detect the coronavirus by utilizing ML and DL algorithms for routine blood tests as an alternative to using the time- and effort-consuming CT scanning technique (255). However, the role of AI was not limited to detection and diagnostic tools. It also contributed to the accurate monitoring of patients during the outbreak of COVID-19. AI was used to automate the contactless scanning process of patients by using data provided by visual sensors such as RGB, Time-of-Flight (TOF) pressure imaging, or thermal cameras (256). In terms of psychological monitoring, researchers further developed previously used ML models to assess the presence of depression and anxiety and predict the most susceptible cases, especially among university students, to avoid more severe cases of mental health decline (257, 258). Furthermore, ML methods (i.e., Support Vector Machine, Artificial Neural Networks, Random Forest, Decision Tree, Logistic Regression, and K-Nearest Neighbor) also contributed to the management of health systems by predicting the clinical characteristics of patients to identify health risks and predicting mortality as researchers relied in some cases on analyzing some clinical criteria such as oxygen level, the genome type, and patient's phenotypic comorbidity, or socio-demographic data to predict the course of the disease (259). The use of AI algorithms has also accelerated drug testing. For instance, a deep learning system created by Google DeepMind (Alpha Fold) provided critical information about protein structures related to COVID-19 for use in vaccine discovery, which would take much longer if implemented through traditional experimental methods (251).

Although the COVID-19 pandemic has successfully accelerated the use of AI applications in the health sector by machine and deep learning models, human skills and laboratory or clinical experiments are still critical to ensure the accuracy of the data provided by AI techniques. It is also notable that, while representing a crucial leap in scientific research, results based on data collection may raise ethical concerns, particularly regarding privacy and discrimination.

Nanotechnology vs. COVID-19

The roll-out of nano-biosensors and vaccines for COVID-19 demonstrates the crucial role of nanotechnology in combating the public health crisis (260, 261).

Nano-biosensors for COVID-19

Diagnostic testing for COVID-19 is typically done with RT-qPCR, serological, or antigen tests. Using nano-biosensors is a cheaper, faster, and more effective method recently developed that can detect viral infection. Nano-biosensor fabrication might be described simply as the physical and/or chemical immobilization of biological molecules such as proteins, antibodies, nucleic acids, enzymes, and cells on the surface of a transducer. This biological component interacts selectively with the counter antibodies, antigens, or proteins, resulting in various changes (e.g., mechanical, electrochemical, thermal, and optical) measured by the transducer. Surface modification with nanoparticles of different shapes and sizes effectively boosts the detection sensitivity and accuracy (260, 262).

Because of their distinctive optical and plasmonic features, gold nanoparticles have piqued the interest of researchers working on COVID-19 nano-biosensor production (263). A gold nanoparticle-based nano-biosensor based on the aggregation behavior of gold nanoparticles was reported for the colorimetric detection of COVID-19 (264). Another nano-biosensor based on 4-aminothiophenol-functionalized gold nanoparticles was developed for quick COVID-19 diagnosis, relying on the interaction of the COVID-19 antigen with the functionalized gold nanoparticles (265). Magnetic nanoparticles, such as negatively charged zinc ferrite nanoparticles, have been discussed for COVID-19 detection by interaction with viral RNA concentration (266). Furthermore, a lateral flow immunoassay kit based on selenium nanoparticles was introduced as a simple, quick test for detecting the COVID-19 virus based on color change (267). On top of that, carbon nanotubes were used to create a variety of smartphone-based COVID-19-detecting biosensors (268, 269). Nano-biosensors are more sensitive, accurate, faster, dependent, and practical for simple applications compared to existing diagnostic techniques for COVID-19 detection (such as RT-PCR, CT imaging, serological immunoassay, etc.) (260, 270272).

Nano-based COVID-19 vaccines

Nanovaccinology proved invaluable in battling the COVID-19 pandemic, supplying the globe with viable vaccine formulations against COVID-19 in an incredible amount of time. Nanotechnology aided in developing nanoparticle platforms for administering molecular vaccines that protect against premature degradation and off-targeting. To obtain stimuli-responsiveness and high immunogenicity, the nanoparticle vaccines might be modified by changing the physicochemical properties, including targeting moieties on their surface and/or co-delivery adjuvants (273276). Particulate antigen approaches and nano adjuvants are used in numerous cutting-edge vaccinations. Generally, the nano-platforms used for vaccination could be summarized as virus-like particles (VLPs), polymeric nanoparticles (PNPs), inorganic nanoparticles, virosomes, and lipid nanoparticles (LNPs).

VLPs, the most popular platform, comprise the virus surface proteins that are highly organized in symmetrical architectures without the genetic material, leading to safe and productive vaccinations (277, 278). The few studies conducted to develop VLPs containing the structural proteins of SARS-CoV-2, such as the spike protein, have shown tolerability and immunogenicity (279, 280).

PNPs are formed by self-assembly of architecturally structured synthetic polymers such as poly lactide-co-glycolic acid or natural proteins. PNPs are utilized as cargo to sustain vaccine release and/or as adjuvants to activate the immune system (281, 282). In that regard, Ufovax's self-assembling protein nanoparticle (1c-SApNP) vaccine platform technology has been exploited to develop a vaccine against SARS-CoV-2 (283).

Inorganic NPs that use gold and silica are advantageous in terms of the high loading capacity of the antigen, which can encapsulate into the mesoporous structures and is stabilized by hydrophobic or electrostatic forces. However, the toxicity, non-biodegradability, and bioaccumulation of the inorganic nanoparticles are questionable (284, 285). In a brief investigation, gold NPs were explored as a nanocarrier platform and adjuvant for COVID-19 immunization, yet the vaccination improvement was poor. (286). On the other hand, biodegradable mesoporous silica nanoparticles were employed as a nanocarrier for ten different epitopes of the spike (S) glycoprotein of SARS-CoV-2, and the in vivo results revealed high activation of particular immune responses (287).

Virosomes are liposomes that are characterized by carrying conjugated antigenic epitopes on their surface. Virosomal vaccines against hepatitis A and influenza viruses are already marketed under the trade names Epaxal® and Inflexal® (288, 289). Despite being able to provide effective vaccination, the trials to create a COVID-19 vaccine candidate based on virosomes were limited (290, 291).

LNPs played a key role in nanovaccinology during the pandemic because they enabled the delivery of the highly effective and affordable COVID-19 vaccine's mRNA (292, 293). LNPs have an added benefit in nanovaccinology because they protect mRNA from phagocyte absorption and endogenous nuclease destruction while also facilitating the diffusion of charged hydrophilic mRNA through hydrophobic cell membranes for the expression of proteins (293, 294). LNPs for mRNA distribution are highly symmetrical, hydrophobic structures made mostly of ionizable cationic lipids, which are employed to circumvent the toxicity disadvantage of regular cationic lipids. The ionizable cationic lipids are distributed primarily in the hydrophilic core, forming internal hydrophobic areas that ionize during acidic manufacturing conditions, allowing electrostatic encapsulation of mRNA. In contrast, these ionizable cationic lipids at physiological pHs become uncharged and thus more biocompatible (293, 295, 296). PEGylated lipids are also employed to cover the core, reducing opsonization and so avoiding uptake by phagocytes, and increasing vaccine circulation within the blood (297, 298). Moderna (mRNA-1273) and Pfizer-BioNTech (BNT162b2) are the two mRNA-based LNP COVID-19 vaccines that have been emergently licensed by the US-FDA to address this crisis (293). Both vaccines are made of ionizable cationic lipid, PEGylated lipid, cholesterol, and distearoylphosphatidylcholine and encapsulate mRNA encoding the SARS-CoV-2 spike protein, which undergoes translation in the cytoplasm into the SARS-CoV-2 spike protein, triggering a reaction by the immune system (273, 299).

Conclusions and future perspectives

The rapid global response to the COVID-19 pandemic has led to significant advancements in our understanding of the virus and its treatment. Researchers and medical professionals have collaborated to identify effective treatments, develop vaccines, and leverage artificial intelligence and nano-based technologies for diagnostic aid. The evolution of COVID-19, from its etiology to pathogenicity and pathophysiology, has been thoroughly explored in recent literature. Both synthetic and natural treatments have been investigated, and the scientific community remains vigilant in addressing the ongoing challenges posed by SARS-CoV-2. As we continue to learn and adapt, the collective efforts of scientists worldwide will play a crucial role in mitigating the impact of COVID-19 and preventing future outbreaks. The COVID-19 pandemic, caused by the positive-strand RNA virus SARS-CoV-2, has presented significant challenges to global health. The virus's new strains, classified into variants of concern and variants of interest, including Alpha, Beta, Gamma, and Delta variants, have further complicated the situation. The etiology of COVID-19 shows that symptoms may be accentuated with comorbidities like age. Additionally, COVID-19 has been associated with acute respiratory disease, acute kidney injury, cardiovascular damage, and other complications. It exacerbates systemic inflammation, which can be detrimental for those with preexisting conditions like coronary heart disease. The virus may also induce multiorgan damage, repression of the immune system, hypercytokinemia, and more.

Various treatments have been implemented from antiviral therapies, corticosteroid therapies, immunotherapies, dietary supplements, and more. Additionally, artificial intelligence, in conjunction with other technologies, has been used to aid in the fight against the virus. Despite the promise this new technology holds for detecting and monitoring infections, it is only in its developing stages and must be further tested before it is fully implemented with regard to other communicable diseases. In the case of pregnant women, there is evidence of greater rates of miscarriage and hypertension, indicating placental involvement. However, the majority of research has found no evidence that the placenta contains SARS-CoV-2 RNA. The latest research indicates that vertical transmission of SARS-CoV-2 is improbable, but pregnant women may be at a higher risk for more severe consequences. As we continue to battle this pandemic, it is crucial to keep researching and developing new strategies and treatments. The collective efforts of scientists worldwide will play a crucial role in mitigating the impact of COVID-19 and preventing future outbreaks. The dynamic association between humoral and cellular immunity during the battle against SARS-CoV-2 infection is an area of interest for future research. The lessons learned from this pandemic will undoubtedly be valuable in preparing for and managing future health crises.

Author contributions

AE: Conceptualization, Data curation, Investigation, Writing – original draft, Writing – review & editing. SE: Conceptualization, Investigation, Methodology, Resources, Visualization, Writing – original draft, Writing – review & editing. RA: Conceptualization, Data curation, Investigation, Methodology, Validation, Writing – original draft, Writing – review & editing. NW: Conceptualization, Data curation, Formal analysis, Investigation, Writing – original draft, Writing – review & editing. AHE: Conceptualization, Investigation, Project administration, Resources, Supervision, Validation, Writing – original draft, Writing – review & editing. AH: Conceptualization, Formal analysis, Project administration, Visualization, Writing – original draft, Writing – review & editing. MM: Conceptualization, Data curation, Formal analysis, Methodology, Software, Writing – original draft, Writing – review & editing. ME: Data curation, Formal analysis, Investigation, Methodology, Writing – original draft, Writing – review & editing. EG: Conceptualization, Formal analysis, Project administration, Visualization, Writing – original draft, Writing – review & editing. RF: Data curation, Project administration, Resources, Validation, Writing – original draft, Writing – review & editing. JB: Conceptualization, Data curation, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing. UA: Conceptualization, Formal analysis, Investigation, Methodology, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing. MZ: Conceptualization, Data curation, Formal analysis, Methodology, Software, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Acknowledgments

Authors thank Prof. Windy Maury for fruitful discussions.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Generative AI statement

The author(s) declare that no Gen AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fmed.2025.1569013/full#supplementary-material

References

1. Woo PC, Lau SK, Lam CS, Lau CC, Tsang AK, Lau JH, et al. Discovery of seven novel Mammalian and avian coronaviruses in the genus deltacoronavirus supports bat coronaviruses as the gene source of alphacoronavirus and betacoronavirus and avian coronaviruses as the gene source of gammacoronavirus and deltacoronavirus. J Virol. (2012) 86:3995–4008. doi: 10.1128/JVI.06540-11

PubMed Abstract | Crossref Full Text | Google Scholar

2. Dudas G, Carvalho LM, Rambaut A, Bedford T. MERS-CoV spillover at the camel-human interface. Elife. (2018) 7. doi: 10.7554/eLife.31257

PubMed Abstract | Crossref Full Text | Google Scholar

3. Corman VM, Muth D, Niemeyer D, Drosten C. Hosts and Sources of Endemic Human Coronaviruses. Adv Virus Res. (2018) 100:163–88. doi: 10.1016/bs.aivir.2018.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

4. Graham RL, Baric RS. Recombination, reservoirs, and the modular spike: mechanisms of coronavirus cross-species transmission. J Virol. (2010) 84:3134–46. doi: 10.1128/JVI.01394-09

PubMed Abstract | Crossref Full Text | Google Scholar

5. Zhou P, Yang XL, Wang XG, Hu B, Zhang L, Zhang W, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. (2020) 579:270–3. doi: 10.1038/s41586-020-2012-7

PubMed Abstract | Crossref Full Text | Google Scholar

6. Tang X, Wu C, Li X, Song Y, Yao X, Wu X, et al. On the origin and continuing evolution of SARS-CoV-2. Natl Sci Rev. (2020) 7:1012–23. doi: 10.1093/nsr/nwaa036

PubMed Abstract | Crossref Full Text | Google Scholar

7. Duffy S, Shackelton LA, Holmes EC. Rates of evolutionary change in viruses: patterns and determinants. Nat Rev Genet. (2008) 9:267–76. doi: 10.1038/nrg2323

PubMed Abstract | Crossref Full Text | Google Scholar

8. Moeller NH, Shi K, Demir Ö, Belica C, Banerjee S, Yin L, et al. Structure and dynamics of SARS-CoV-2 proofreading exoribonuclease ExoN. Proc Natl Acad Sci USA. (2022) 119:e2106379119. doi: 10.1073/pnas.2106379119

PubMed Abstract | Crossref Full Text | Google Scholar

9. Day T, Gandon S, Lion S, Otto SP. On the evolutionary epidemiology of SARS-CoV-2. Curr Biol. (2020) 30:R849–57. doi: 10.1016/j.cub.2020.06.031

PubMed Abstract | Crossref Full Text | Google Scholar

10. Tegally H, Wilkinson E, Giovanetti M, Iranzadeh A, Fonseca V, Giandhari J, et al. Detection of a SARS-CoV-2 variant of concern in South Africa. Nature. (2021) 592:438–43. doi: 10.1038/s41586-021-03402-9

PubMed Abstract | Crossref Full Text | Google Scholar

11. Funk M, Ash C. Evolution and epidemic spread of SARS-CoV-2 in Brazil. Science. (2020) 369:1255–60. doi: 10.1126/science.369.6508.1203-o

Crossref Full Text | Google Scholar

12. Li X, Zai J, Zhao Q, Nie Q, Li Y, Foley BT, et al. Evolutionary history, potential intermediate animal host, and cross-species analyses of SARS-CoV-2. J Med Virol. (2020) 92:602–11. doi: 10.1002/jmv.25731

PubMed Abstract | Crossref Full Text | Google Scholar

13. Zhao Z, Li H, Wu X, Zhong Y, Zhang K, Zhang YP, et al. Moderate mutation rate in the SARS coronavirus genome and its implications. BMC Evol Biol. (2004) 4:21. doi: 10.1186/1471-2148-4-21

PubMed Abstract | Crossref Full Text | Google Scholar

14. Boni MF, Lemey P, Jiang X, Lam TT, Perry BW, Castoe TA, et al. Evolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemic. Nat Microbiol. (2020) 5:1408–17. doi: 10.1038/s41564-020-0771-4

PubMed Abstract | Crossref Full Text | Google Scholar

15. Lam TT, Jia N, Zhang YW, Shum MH, Jiang JF, Zhu HC, et al. Identifying SARS-CoV-2-related coronaviruses in Malayan pangolins. Nature. (2020) 583:282–5. doi: 10.1038/s41586-020-2169-0

PubMed Abstract | Crossref Full Text | Google Scholar

16. Lai MM, Cavanagh D. The molecular biology of coronaviruses. Adv Virus Res. (1997) 48:1–100. doi: 10.1016/S0065-3527(08)60286-9

PubMed Abstract | Crossref Full Text | Google Scholar

17. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat Med. (2020) 26:450–2. doi: 10.1038/s41591-020-0820-9

PubMed Abstract | Crossref Full Text | Google Scholar

18. Mavian C, Pond SK, Marini S, Magalis BR, Vandamme AM, Dellicour S, et al. Sampling bias and incorrect rooting make phylogenetic network tracing of SARS-COV-2 infections unreliable. Proc Natl Acad Sci USA. (2020) 117:12522–3. doi: 10.1073/pnas.2007295117

PubMed Abstract | Crossref Full Text | Google Scholar

19. Korber B, Fischer WM, Gnanakaran S, Yoon H, Theiler J, Abfalterer W, et al. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases infectivity of the COVID-19 virus. Cell. (2020) 182:812–7 e19. doi: 10.1016/j.cell.2020.06.043

PubMed Abstract | Crossref Full Text | Google Scholar

20. Faria NR, Mellan TA, Whittaker C, Whittaker C, Candido DDS, Mishra S, et al. Genomics and epidemiology of the P1 SARS-CoV-2 lineage in Manaus, Brazil. Science. (2021) 372:815–21. doi: 10.1126/science.abh2644

PubMed Abstract | Crossref Full Text | Google Scholar

21. Aiyar Y, Chandru V, Chatterjee M, Desai S, Fernandez A, Gupta A, et al. India's resurgence of COVID-19: urgent actions needed. Lancet. (2021) 397:2232–4. doi: 10.1016/S0140-6736(21)01202-2

PubMed Abstract | Crossref Full Text | Google Scholar

22. COVID-19 Genomics UK (COG-UK) Y29uc29ydGl1bWNvbnRhY3RAY29nY29uc29ydGl1bS51aw==. An integrated national scale SARS-CoV-2 genomic surveillance network. Lancet Microbe. (2020) 1:e99–100. doi: 10.1016/S2666-5247(20)30054-9

PubMed Abstract | Crossref Full Text | Google Scholar

23. Konings F, Perkins MD, Kuhn JH, Pallen MJ, Alm EJ, Archer BN, et al. SARS-CoV-2 Variants of Interest and Concern naming scheme conducive for global discourse. Nat Microbiol. (2021) 6:821–3. doi: 10.1038/s41564-021-00932-w

PubMed Abstract | Crossref Full Text | Google Scholar

24. Karim SSA, Karim QA. Omicron SARS-CoV-2 variant: a new chapter in the COVID-19 pandemic. Lancet. (2021) 398:2126–8. doi: 10.1016/S0140-6736(21)02758-6

PubMed Abstract | Crossref Full Text | Google Scholar

25. Pulliam JRC, van Schalkwyk C, Govender N, von Gottberg A, Cohen C, Groome MJ, et al. Increased risk of SARS-CoV-2 reinfection associated with emergence of Omicron in South Africa. Science. (2022) 376:eabn4947. doi: 10.1126/science.abn4947

PubMed Abstract | Crossref Full Text | Google Scholar

26. Harris M, Hart J, Bhattacharya O, Russell FM. Risk factors for SARS-CoV-2 infection during the early stages of the COVID-19 pandemic: a systematic literature review. Front Public Health. (2023) 11:1178167. doi: 10.3389/fpubh.2023.1178167

PubMed Abstract | Crossref Full Text | Google Scholar

27. Patil NR, Herc ES, Girgis M. Cold Agglutinin disease and autoimmune hemolytic anemia with pulmonary embolism as a presentation of COVID-19 infection. Hematol Oncol Stem Cell Ther. (2022) 15:213–6. doi: 10.1016/j.hemonc.2020.06.005

PubMed Abstract | Crossref Full Text | Google Scholar

28. Mohamadi Yarijani Z, Najafi H. Kidney injury in COVID-19 patients, drug development and their renal complications: review study. Biomed Pharmacother. (2021) 142:111966. doi: 10.1016/j.biopha.2021.111966

PubMed Abstract | Crossref Full Text | Google Scholar

29. Jayaswal SK, Singh S, Malik PS, Venigalla SK, Gupta P, Samaga SN, et al. Detrimental effect of diabetes and hypertension on the severity and mortality of COVID-19 infection: a multi-center case-control study from India. Diabetes Metab Syndr. (2021) 15:102248. doi: 10.1016/j.dsx.2021.102248

PubMed Abstract | Crossref Full Text | Google Scholar

30. Fedeli U, Amidei CB, Casotto V, Schievano E, Zoppini G. Excess diabetes-related deaths: the role of comorbidities through different phases of the COVID-19 pandemic. Nutr Metab Cardiovasc Dis. (2023) 33:1709–15. doi: 10.1016/j.numecd.2023.05.028

PubMed Abstract | Crossref Full Text | Google Scholar

31. Cortés OL, Paipa MDP, Mojica C, Rojas YA, Pulido S, Arias M, et al. Characteristics, treatment, and nursing care of patients infected by Sars-CoV-2 hospitalized in intensive care units: multicenter study of colombian hospitals. Invest Educ Enferm. (2022) 40:e08. doi: 10.17533/udea.iee.v40n1e08

PubMed Abstract | Crossref Full Text | Google Scholar

32. Zhang G, Hu C, Luo L, Fang F, Chen Y, Li J, et al. Clinical features and short-term outcomes of 221 patients with COVID-19 in Wuhan, China. J Clin Virol. (2020) 127:104364. doi: 10.1016/j.jcv.2020.104364

PubMed Abstract | Crossref Full Text | Google Scholar

33. Pilato MS, Urban A, Alkawadri R, Barot NV, Castellano JF, Rajasekaran V, et al. EEG findings in coronavirus disease. J Clin Neurophysiol. (2022) 39:159–65. doi: 10.1097/WNP.0000000000000752

PubMed Abstract | Crossref Full Text | Google Scholar

34. Kreutmair S, Kauffmann M, Unger S, Ingelfinger F, Núñez NG, Alberti C, et al. Preexisting comorbidities shape the immune response associated with severe COVID-19. J Allergy Clin Immunol. (2022) 150:312–24. doi: 10.1016/j.jaci.2022.05.019

PubMed Abstract | Crossref Full Text | Google Scholar

35. Sarioglu E, Sarialtin SY, Çoban T. Neurological complications and effects of COVID-19: symptoms and conceivable mechanisms. Brain Hemorrhages. (2023) 4:154–73. doi: 10.1016/j.hest.2023.02.001

PubMed Abstract | Crossref Full Text | Google Scholar

36. Staico MF, Zaffanello M, DI Pietro G, Fanos V, Marcialis MA. The kidney in COVID-19: protagonist or figurant? Panminerva Med. (2023) 65:65–75. doi: 10.23736/S0031-0808.20.03965-8

PubMed Abstract | Crossref Full Text | Google Scholar

37. Nirenberg MS, Herrera MDMR. Foot manifestations in a patient with COVID-19 and Epstein-Barr virus: a case study. Foot. (2021) 46:101707. doi: 10.1016/j.foot.2020.101707

PubMed Abstract | Crossref Full Text | Google Scholar

38. Bashir H, Yildiz M, Cafardi J, Bhatia A, Garcia S, Henry TD, et al. A review of heart failure in patients with COVID-19. Heart Fail Clin. (2023) 19:e1–8. doi: 10.1016/j.hfc.2023.03.002

PubMed Abstract | Crossref Full Text | Google Scholar

39. Lin L, Lu L, Cao W, Li T. Hypothesis for potential pathogenesis of SARS-CoV-2 infection–a review of immune changes in patients with viral pneumonia. Emerg Microbes Infect. (2020) 9:727–32. doi: 10.1080/22221751.2020.1746199

PubMed Abstract | Crossref Full Text | Google Scholar

40. Navas-Martín S, Weiss SR. Coronavirus replication and pathogenesis: implications for the recent outbreak of severe acute respiratory syndrome (SARS), and the challenge for vaccine development. J Neurovirol. (2004) 10:75–85. doi: 10.1080/13550280490280292

PubMed Abstract | Crossref Full Text | Google Scholar

41. Hanley B, Lucas SB, Youd E, Swift B, Osborn M. Autopsy in suspected COVID-19 cases. J Clin Pathol. (2020) 73:239–42. doi: 10.1136/jclinpath-2020-206522

PubMed Abstract | Crossref Full Text | Google Scholar

42. Zou L, Ruan F, Huang M, Liang L, Huang H, Hong Z, et al. SARS-CoV-2 viral load in upper respiratory specimens of infected patients. N Engl J Med. (2020) 382:1177–9. doi: 10.1056/NEJMc2001737

PubMed Abstract | Crossref Full Text | Google Scholar

43. Li C, He Q, Qian H, Liu J. Overview of the pathogenesis of COVID-19 (Review). Exp Ther Med. (2021) 22:1011. doi: 10.3892/etm.2021.10444

PubMed Abstract | Crossref Full Text | Google Scholar

44. Kattula B, Reddi B, Jangam A, Naik L, Adimoolam BM, Vavilapalli S, et al. Development of 2-chloroquinoline based heterocyclic frameworks as dual inhibitors of SARS-CoV-2 M. Int J Biol Macromol. (2023) 242(Pt 2):124772. doi: 10.1016/j.ijbiomac.2023.124772

PubMed Abstract | Crossref Full Text | Google Scholar

45. Pathania S, Rawal RK, Singh PK. RdRp (RNA-dependent RNA polymerase): a key target providing anti-virals for the management of various viral diseases. J Mol Struct. (2022) 1250:131756. doi: 10.1016/j.molstruc.2021.131756

PubMed Abstract | Crossref Full Text | Google Scholar

46. Jiang MD, Zu ZY, Schoepf UJ, Savage RH, Zhang XL, Lu GM, et al. Current status of etiology, epidemiology, clinical manifestations and imagings for COVID-19. Korean J Radiol. (2020) 21:1138–49. doi: 10.3348/kjr.2020.0526

PubMed Abstract | Crossref Full Text | Google Scholar

47. Fallah A, Sedighian H, Behzadi E, Havaei SA, Kachuei R, Imani Fooladi AA. The role of serum circulating microbial toxins in severity and cytokine storm of COVID positive patients. Microb Pathog. (2023) 174:105888. doi: 10.1016/j.micpath.2022.105888

PubMed Abstract | Crossref Full Text | Google Scholar

48. Nuccetelli M, Pieri M, Gisone F, Bernardini S. Combined anti-SARS-CoV-2 IgA, IgG, and IgM detection as a better strategy to prevent second infection spreading waves. Immunol Invest. (2022) 51:233–45. doi: 10.1080/08820139.2020.1823407

PubMed Abstract | Crossref Full Text | Google Scholar

49. Rauf A, Abu-Izneid T, Olatunde A, Ahmed Khalil A, Alhumaydhi FA, Tufail T, et al. COVID-19 pandemic: epidemiology, etiology, conventional and non-conventional therapies. Int J Environ Res Public Health. (2020) 17:8155. doi: 10.3390/ijerph17218155

PubMed Abstract | Crossref Full Text | Google Scholar

50. Oudit GY, Wang K, Viveiros A, Kellner MJ, Penninger JM. Angiotensin-converting enzyme 2-at the heart of the COVID-19 pandemic. Cell. (2023) 186:906–22. doi: 10.1016/j.cell.2023.01.039

PubMed Abstract | Crossref Full Text | Google Scholar

51. Nemati Zargaran F, Rostamian M, Kooti S, Madanchi H, Ghadiri K. Co-infection of COVID-19 and parasitic diseases: a systematic review. Parasite Epidemiol Control. (2023) 21:e00299. doi: 10.1016/j.parepi.2023.e00299

PubMed Abstract | Crossref Full Text | Google Scholar

52. Hamming I, Timens W, Bulthuis ML, Lely AT, Navis G, van Goor H. Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus A first step in understanding SARS pathogenesis. J Pathol. (2004) 203:631–7. doi: 10.1002/path.1570

PubMed Abstract | Crossref Full Text | Google Scholar

53. Dahal A, Sonju JJ, Kousoulas KG, Jois SD. Peptides and peptidomimetics as therapeutic agents for COVID-19. Pept Sci. (2022) 114:e24245. doi: 10.1002/pep2.24245

PubMed Abstract | Crossref Full Text | Google Scholar

54. Paniri A, Hosseini MM, Moballegh-Eslam M, Akhavan-Niaki H. Comprehensive in silico identification of impacts of. Gene Rep. (2021) 22:100979. doi: 10.1016/j.genrep.2020.100979

PubMed Abstract | Crossref Full Text | Google Scholar

55. Koch J, Uckeley ZM, Doldan P, Stanifer M, Boulant S, Lozach PY. TMPRSS2 expression dictates the entry route used by SARS-CoV-2 to infect host cells. Embo J. (2021) 40:e107821. doi: 10.15252/embj.2021107821

PubMed Abstract | Crossref Full Text | Google Scholar

56. Arman K, Dalloul Z, Bozgeyik E. Emerging role of microRNAs and long non-coding RNAs in COVID-19 with implications to therapeutics. Gene. (2023) 861:147232. doi: 10.1016/j.gene.2023.147232

PubMed Abstract | Crossref Full Text | Google Scholar

57. van der Poll T, Opal SM. Host–pathogen interactions in sepsis. Lancet Infect Dis. (2008) 8:32–43. doi: 10.1016/S1473-3099(07)70265-7

PubMed Abstract | Crossref Full Text | Google Scholar

58. Streicher F, Jouvenet N. Stimulation of innate immunity by host and viral RNAs. Trends Immunol. (2019) 40:1134–48. doi: 10.1016/j.it.2019.10.009

PubMed Abstract | Crossref Full Text | Google Scholar

59. Fu Y, Cheng Y, Wu Y. Understanding SARS-CoV-2-mediated inflammatory responses: from mechanisms to potential therapeutic tools. Virol Sin. (2020) 35:266–71. doi: 10.1007/s12250-020-00207-4

PubMed Abstract | Crossref Full Text | Google Scholar

60. Eppensteiner J, Kwun J, Scheuermann U, Barbas A, Limkakeng AT, Kuchibhatla M, et al. Damage-and pathogen-associated molecular patterns play differential roles in late mortality after critical illness. JCI Insight. (2019) 4:e127925. doi: 10.1172/jci.insight.127925

PubMed Abstract | Crossref Full Text | Google Scholar

61. Chua RL, Lukassen S, Trump S, Hennig BP, Wendisch D, Pott F, et al. COVID-19 severity correlates with airway epithelium–immune cell interactions identified by single-cell analysis. Nat Biotechnol. (2020) 38:970–9. doi: 10.1038/s41587-020-0602-4

PubMed Abstract | Crossref Full Text | Google Scholar

62. Ren X, Wen W, Fan X, Hou W, Su B, Cai P, et al. COVID-19 immune features revealed by a large-scale single-cell transcriptome atlas. Cell. (2021) 184:1895–913. e19.

Google Scholar

63. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. (2020) 8:420–2. doi: 10.1016/S2213-2600(20)30076-X

PubMed Abstract | Crossref Full Text | Google Scholar

64. Diao B, Wang C, Tan Y, Chen X, Liu Y, Ning L, et al. Reduction and functional exhaustion of T cells in patients with coronavirus disease (2019) (COVID-19) Front Immunol. (2020) 2020:827. doi: 10.3389/fimmu.2020.00827

PubMed Abstract | Crossref Full Text | Google Scholar

65. Swerdlow DL, Finelli L. Preparation for possible sustained transmission of 2019 novel coronavirus: lessons from previous epidemics. JAMA. (2020) 323:1129–30. doi: 10.1001/jama.2020.1960

PubMed Abstract | Crossref Full Text | Google Scholar

66. Tian X, Li C, Huang A, Xia S, Lu S, Shi Z, et al. Potent binding of 2019 novel coronavirus spike protein by a SARS coronavirus-specific human monoclonal antibody. Emerg Microbes Infect. (2020) 9:382–5. doi: 10.1080/22221751.2020.1729069

PubMed Abstract | Crossref Full Text | Google Scholar

67. Ni L, Ye F, Cheng ML, Feng Y, Deng YQ, Zhao H, et al. Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity. (2020) 52:971–7.e3. doi: 10.1016/j.immuni.2020.04.023

PubMed Abstract | Crossref Full Text | Google Scholar

68. Chen T, Wu D, Chen H, Yan W, Yang D, Chen G, et al. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ. (2020) 368:m1295. doi: 10.1136/bmj.m1295

PubMed Abstract | Crossref Full Text | Google Scholar

69. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. (2020) 8:475–81. doi: 10.1016/S2213-2600(20)30079-5

PubMed Abstract | Crossref Full Text | Google Scholar

70. Channappanavar R, Perlman S. Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology. Semin Immunopathol. (2017) 39:529–39. doi: 10.1007/s00281-017-0629-x

PubMed Abstract | Crossref Full Text | Google Scholar

71. Zhou J, Chu H, Li C, Wong BH, Cheng ZS, Poon VK, et al. Active replication of Middle East respiratory syndrome coronavirus and aberrant induction of inflammatory cytokines and chemokines in human macrophages: implications for pathogenesis. J Infect Dis. (2014) 209:1331–42. doi: 10.1093/infdis/jit504

PubMed Abstract | Crossref Full Text | Google Scholar

72. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Investig. (2020) 130:2620–9. doi: 10.1172/JCI137244

PubMed Abstract | Crossref Full Text | Google Scholar

73. Misra DP, Agarwal V, Gasparyan AY, Zimba O. Rheumatologists' perspective on coronavirus disease 19 (COVID-19) and potential therapeutic targets. Clin Rheumatol. (2020) 39:2055–62. doi: 10.1007/s10067-020-05073-9

PubMed Abstract | Crossref Full Text | Google Scholar

74. Padureanu R, Albu CV, Mititelu RR, Bacanoiu MV, Docea AO, Calina D, et al. Oxidative stress and inflammation interdependence in multiple sclerosis. J Clinic Med. (2019) 8:1815. doi: 10.3390/jcm8111815

PubMed Abstract | Crossref Full Text | Google Scholar

75. Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. (2020) 382:1708–20. doi: 10.1056/NEJMOA2002032

PubMed Abstract | Crossref Full Text | Google Scholar

76. Langford BJ, So M, Raybardhan S, Leung V, Westwood D, MacFadden DR, et al. Bacterial co-infection and secondary infection in patients with COVID-19: a living rapid review and meta-analysis. Clin Microbiol Infect. (2020) 26:1622–9. doi: 10.1016/j.cmi.2020.07.016

PubMed Abstract | Crossref Full Text | Google Scholar

77. Liang W, Guan W, Chen R, Wang W, Li J, Xu K, et al. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol. (2020) 21:335–7. doi: 10.1016/S1470-2045(20)30096-6

PubMed Abstract | Crossref Full Text | Google Scholar

78. Mason RJ. Thoughts on the alveolar phase of COVID-19. Am J Physiol-Lung Cell Mol Physiol. (2020) 319:L115–20. doi: 10.1152/ajplung.00126.2020

PubMed Abstract | Crossref Full Text | Google Scholar

79. Greenhalgh T, Jimenez JL, Prather KA, Tufekci Z, Fisman D, Schooley R. Ten scientific reasons in support of airborne transmission of SARS-CoV-2. Lancet. (2021) 397:1603–5. doi: 10.1016/S0140-6736(21)00869-2

PubMed Abstract | Crossref Full Text | Google Scholar

80. Alon R, Sportiello M, Kozlovski S, Kumar A, Reilly EC, Zarbock A, et al. Leukocyte trafficking to the lungs and beyond: lessons from influenza for COVID-19. Nat Rev Immunol. (2021) 21:49–64. doi: 10.1038/s41577-020-00470-2

PubMed Abstract | Crossref Full Text | Google Scholar

81. Erdinc B, Sahni S, Gotlieb V. Hematological manifestations and complications of COVID-19. Adv Clin Exp Med. (2021) 30:101–7. doi: 10.17219/acem/130604

PubMed Abstract | Crossref Full Text | Google Scholar

82. Spottiswoode N, Tsitsiklis A, Chu VT, Phan HV, DeVoe C, Love C, et al. Microbial dynamics and pulmonary immune responses in COVID-19 secondary bacterial pneumonia. Nat Commun. (2024) 15:9339. doi: 10.1038/s41467-024-53566-x

PubMed Abstract | Crossref Full Text | Google Scholar

83. Leffler CT, Lykins V JD, Das S, Yang E, Konda S. Preliminary analysis of excess mortality in India during the COVID-19 pandemic. Am J Trop Med Hyg. (2022) 106: 1507. doi: 10.4269/ajtmh.21-0864

PubMed Abstract | Crossref Full Text | Google Scholar

84. Godoy LC, Goligher EC, Lawler PR, Slutsky AS, Zarychanski R. Anticipating and managing coagulopathy and thrombotic manifestations of severe COVID-19. CMAJ. (2020) 192:E1156–61. doi: 10.1503/cmaj.201240

PubMed Abstract | Crossref Full Text | Google Scholar

85. Delorey TM, Ziegler CGK, Heimberg G, Normand R, Yang Y, Segerstolpe Å, et al. COVID-19 tissue atlases reveal SARS-CoV-2 pathology and cellular targets. Nature. (2021) 595:107–13. doi: 10.1038/s41586-021-03570-8

PubMed Abstract | Crossref Full Text | Google Scholar

86. Meleti M, Cassi D, Bueno L, Bologna-Molina R. COVID-19 diffusion and its impact on dental practice in distant countries with similar ethnic background. Oral Dis. (2021) 27(Suppl. 3):720–2. doi: 10.1111/odi.13438

PubMed Abstract | Crossref Full Text | Google Scholar

87. Huang N, Pérez P, Kato T, Mikami Y, Okuda K, Gilmore RC, et al. SARS-CoV-2 infection of the oral cavity and saliva. Nat Med. (2021) 27:892–903. doi: 10.1038/s41591-021-01296-8

PubMed Abstract | Crossref Full Text | Google Scholar

88. Marouf N, Cai W, Said KN, Daas H, Diab H, Chinta VR, et al. Association between periodontitis and severity of COVID-19 infection: a case–control study. J Clin Periodontol. (2021) 48:483–91. doi: 10.1111/jcpe.13435

PubMed Abstract | Crossref Full Text | Google Scholar

89. Chan SL, Kudo M. Impacts of COVID-19 on Liver cancers: during and after the pandemic. Liver Cancer. (2020) 9:491–502. doi: 10.1159/000510765

PubMed Abstract | Crossref Full Text | Google Scholar

90. Gracia-Ramos AE, Jaquez-Quintana JO, Contreras-Omaña R, Auron M. Liver dysfunction and SARS-CoV-2 infection. World J Gastroenterol. (2021) 27:3951–70. doi: 10.3748/wjg.v27.i26.3951

PubMed Abstract | Crossref Full Text | Google Scholar

91. Mikolasevic I, Bozic D, Pavić T, Ruzic A, Hauser G, Radic M, et al. Liver disease in the era of COVID-19: is the worst yet to come? World J Gastroenterol. (2021) 27:6039–52. doi: 10.3748/wjg.v27.i36.6039

PubMed Abstract | Crossref Full Text | Google Scholar

92. Tapper EB, Asrani SK. The COVID-19 pandemic will have a long-lasting impact on the quality of cirrhosis care. J Hepatol. (2020) 73:441–5. doi: 10.1016/j.jhep.2020.04.005

PubMed Abstract | Crossref Full Text | Google Scholar

93. Iavarone M, Sangiovanni A, Carrafiello G, Rossi G, Lampertico P. Management of hepatocellular carcinoma in the time of COVID-19. Ann Oncol. (2020) 31:1084–5. doi: 10.1016/j.annonc.2020.04.007

PubMed Abstract | Crossref Full Text | Google Scholar

94. Guo M, Tao W, Flavell RA, Zhu S. Potential intestinal infection and faecal–oral transmission of SARS-CoV-2. Nat Rev Gastroenterol Hepatol. (2021) 18:269–83. doi: 10.1038/s41575-021-00416-6

PubMed Abstract | Crossref Full Text | Google Scholar

95. Yeoh YK, Zuo T, Lui GC, Zhang F, Liu Q, Li AY, et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. (2021) 70:698–706. doi: 10.1136/gutjnl-2020-323020

PubMed Abstract | Crossref Full Text | Google Scholar

96. Krasemann S, Haferkamp U, Pfefferle S, Woo MS, Heinrich F, Schweizer M, et al. The blood-brain barrier is dysregulated in COVID-19 and serves as a CNS entry route for SARS-CoV-2. Stem Cell Rep. (2022) 17:307–20. doi: 10.1016/j.stemcr.2021.12.011

PubMed Abstract | Crossref Full Text | Google Scholar

97. Marzano AV, Cassano N, Genovese G, Moltrasio C, Vena GA. Cutaneous manifestations in patients with COVID-19: a preliminary review of an emerging issue. Br J Dermatol. (2020) 183:431–42. doi: 10.1111/bjd.19264

PubMed Abstract | Crossref Full Text | Google Scholar

98. Magro CM, Mulvey J, Kubiak J, Mikhail S, Suster D, Crowson AN, et al. Severe COVID-19: a multifaceted viral vasculopathy syndrome. Ann Diagn Pathol. (2021) 50:151645. doi: 10.1016/j.anndiagpath.2020.151645

PubMed Abstract | Crossref Full Text | Google Scholar

99. Trippella G, Ciarcià M, Ferrari M, Buzzatti C, Maccora I, Azzari C, et al. COVID-19 in pregnant women and neonates: a systematic review of the literature with quality assessment of the studies. Pathogens. (2020) 9:485. doi: 10.3390/pathogens9060485

PubMed Abstract | Crossref Full Text | Google Scholar

100. Bohn MK, Hall A, Sepiashvili L, Jung B, Steele S, Adeli K. Pathophysiology of COVID-19: mechanisms underlying disease severity and progression. Physiology. (2020) 35:288–301. doi: 10.1152/physiol.00019.2020

PubMed Abstract | Crossref Full Text | Google Scholar

101. Banks WA, Farrell CL. Impaired transport of leptin across the blood-brain barrier in obesity is acquired and reversible. Am J Physiol-Endocrinol Metab. (2003) 285:E10–5. doi: 10.1152/ajpendo.00468.2002

PubMed Abstract | Crossref Full Text | Google Scholar

102. Henry BM, Benoit SW, de Oliveira MHS, Hsieh WC, Benoit J, Ballout RA, et al. Laboratory abnormalities in children with mild and severe coronavirus disease 2019 (COVID-19): a pooled analysis and review. Clin Biochem. (2020) 81:1–8. doi: 10.1016/j.clinbiochem.2020.05.012

PubMed Abstract | Crossref Full Text | Google Scholar

103. Soper D. Trichomoniasis: under control or undercontrolled? Am J Obstet Gynecol. (2004) 190:281–90. doi: 10.1016/j.ajog.2003.08.023

PubMed Abstract | Crossref Full Text | Google Scholar

104. Vickers NJ. Animal communication: when i'm calling you, will you answer too? Curr Biol. (2017) 27:R713–5. doi: 10.1016/j.cub.2017.05.064

PubMed Abstract | Crossref Full Text | Google Scholar

105. Li G, Hilgenfeld R, Whitley R, De Clercq E. Therapeutic strategies for COVID-19: progress and lessons learned. Nat Rev Drug Discov. (2023) 22:449–75. doi: 10.1038/s41573-023-00672-y

PubMed Abstract | Crossref Full Text | Google Scholar

106. Toussi SS, Hammond JL, Gerstenberger BS, Anderson AS. Therapeutics for COVID-19. Nat Microbiol. (2023) 8:771–86. doi: 10.1038/s41564-023-01356-4

PubMed Abstract | Crossref Full Text | Google Scholar

107. World Health Organization. Therapeutics and COVID-19: Living Guideline. Geneva: World Health Organization (2022).

Google Scholar

108. Ikeda S, Misumi T, Izumi S, Sakamoto K, Nishimura N, Ro S, et al. Corticosteroids for hospitalized patients with mild to critically-ill COVID-19: a multicenter, retrospective, propensity score-matched study. Sci Rep. (2021) 11:1–13. doi: 10.1038/s41598-021-90246-y

PubMed Abstract | Crossref Full Text | Google Scholar

109. National Institutes of Health. COVID-19 Treatment Guidelines Panel. Bethesda, MD: National Institutes of Health (US) (2021).

Google Scholar

110. RECOVERY Collaborative Group, RECOVERY Collaborative Group. Higher dose corticosteroids in patients admitted to hospital with COVID-19 who are hypoxic but not requiring ventilatory support (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. (2023) 401:1499–507. doi: 10.1016/S0140-6736(23)00510-X

PubMed Abstract | Crossref Full Text | Google Scholar

111. World Health Organization. Clinical Management of COVID-19: Living Guideline. Geneva: World Health Organization (2023).

Google Scholar

112. van de Veerdonk FL, Giamarellos-Bourboulis E, Pickkers P, Derde L, Leavis H, van Crevel R, et al. A guide to immunotherapy for COVID-19. Nat Med. (2022) 28:39–50. doi: 10.1038/s41591-021-01643-9

PubMed Abstract | Crossref Full Text | Google Scholar

113. Alijotas-Reig J, Esteve-Valverde E, Belizna C, Selva-O'Callaghan A, Pardos-Gea J, Quintana A, et al. Immunomodulatory therapy for the management of severe COVID-19 Beyond the anti-viral therapy: a comprehensive review. Autoimmun Rev. (2020) 19:102569. doi: 10.1016/j.autrev.2020.102569

PubMed Abstract | Crossref Full Text | Google Scholar

114. Rizk JG, Kalantar-Zadeh K, Mehra MR, Lavie CJ, Rizk Y, Forthal DN. Pharmaco-immunomodulatory therapy in COVID-19. Drugs. (2020) 80:1267–92. doi: 10.1007/s40265-020-01367-z

PubMed Abstract | Crossref Full Text | Google Scholar

115. Peter N, Kerschbaumer A, Dörner T, Dougados M, Fleischmann RM, Geissler K, et al. Points to consider for the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: a consensus statement. Ann Rheum Dis. (2021) 80:71. doi: 10.1136/annrheumdis-2020-218398

PubMed Abstract | Crossref Full Text | Google Scholar

116. Marconi VC, Ramanan AV, de Bono S, Kartman CE, Krishnan V, Liao R, et al. Efficacy and safety of baricitinib for the treatment of hospitalised adults with COVID-19 (COV-BARRIER): a randomised, double-blind, parallel-group, placebo-controlled phase 3 trial. Lancet Respir Med. (2021) 9:1407–18. doi: 10.1016/S2213-2600(21)00331-3

PubMed Abstract | Crossref Full Text | Google Scholar

117. Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic treatments for coronavirus disease (2019) (COVID-19): a review. JAMA. (2020) 323:1824–36. doi: 10.1001/jama.2020.6019

PubMed Abstract | Crossref Full Text | Google Scholar

118. Tummino TA, Rezelj VV, Fischer B, Fischer A, O'Meara MJ, Monel B, et al. Drug-induced phospholipidosis confounds drug repurposing for SARS-CoV-2. Science. (2021) 373:541–7. doi: 10.1126/science.abi4708

PubMed Abstract | Crossref Full Text | Google Scholar

119. Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. (2020) 30:269–71. doi: 10.1038/s41422-020-0282-0

PubMed Abstract | Crossref Full Text | Google Scholar

120. Ferner, R.E. and J.K. Aronson, Chloroquine and hydroxychloroquine in COVID-19. BMJ. (2020) 369:m1432. doi: 10.1136/bmj.m1432

PubMed Abstract | Crossref Full Text | Google Scholar

121. Touret F, de Lamballerie X. Of chloroquine and COVID-19. Antiviral Res. (2020) 177:104762. doi: 10.1016/j.antiviral.2020.104762

PubMed Abstract | Crossref Full Text | Google Scholar

122. Hoffmann M, Mösbauer K, Hofmann-Winkler H, Kaul A, Kleine-Weber H, Krüger N, et al. Chloroquine does not inhibit infection of human lung cells with SARS-CoV-2. Nature. (2020) 585:588–90. doi: 10.1038/s41586-020-2575-3

PubMed Abstract | Crossref Full Text | Google Scholar

123. Diesendorf V, Roll V, Geiger N, Fähr S, Obernolte H, Sewald K, et al. Drug-induced phospholipidosis is not correlated with the inhibition of SARS-CoV-2 - inhibition of SARS-CoV-2 is cell line-specific. Front Cell Infect Microbiol. (2023) 13:1100028. doi: 10.3389/fcimb.2023.1100028

PubMed Abstract | Crossref Full Text | Google Scholar

124. Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease (2019) (COVID-19): a review. JAMA. (2020) 324:782–93. doi: 10.1001/jama.2020.12839

PubMed Abstract | Crossref Full Text | Google Scholar

125. Self WH, Semler MW, Leither LM, Casey JD, Angus DC, Brower RG, et al. Effect of hydroxychloroquine on clinical status at 14 days in Hospitalized patients with COVID-19: a randomized clinical trial. JAMA. (2020) 324:2165–76. doi: 10.1001/jama.2020.22240

PubMed Abstract | Crossref Full Text | Google Scholar

126. World Health Organization. Drugs to Prevent COVID-19: Living Guideline. Geneva: World Health Organization (2023).

Google Scholar

127. 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) 18:1094–9. doi: 10.1111/jth.14817

PubMed Abstract | Crossref Full Text | Google Scholar

128. Kattakola Y, Prasad R, Sharma R, Wanjari MB. High-dose prophylactic anticoagulation for COVID-19 pneumonia: a review of benefits and risks. Cureus. (2023) 15:e37705. doi: 10.7759/cureus.37705

PubMed Abstract | Crossref Full Text | Google Scholar

129. Zhou B, Kojima S, Kawamoto A, Fukushima M. COVID-19 pathogenesis, prognostic factors, and treatment strategy: urgent recommendations. J Med Virol. (2021) 93:2694–704. doi: 10.1002/jmv.26754

PubMed Abstract | Crossref Full Text | Google Scholar

130. The Office of Dietary Supplements (ODS). Dietary Supplements in the Time of COVID-19. Bethesda, MD: The National Institutes of Health (NIH) (2023).

Google Scholar

131. Zhang L, Liu Y. Potential interventions for novel coronavirus in China: a systematic review. J Med Virol. (2020) 92:479–90. doi: 10.1002/jmv.25707

PubMed Abstract | Crossref Full Text | Google Scholar

132. Cundra LB, Vallabhaneni M, Saadeh M, Houston KV, Yoo BS, D'Souza S, et al. Immunomodulation strategies against COVID-19 evidence: key nutrients and dietary approaches. Explor Med. (2023) 4:189–206. doi: 10.37349/emed.2023.00133

PubMed Abstract | Crossref Full Text | Google Scholar

133. Iddir M, Brito A, Dingeo G, Fernandez Del Campo SS, Samouda H, La Frano MR, et al. Strengthening the immune system and reducing inflammation and oxidative stress through diet and nutrition: considerations during the COVID-19 crisis. Nutrients. (2020) 12:1562. doi: 10.3390/nu12061562

PubMed Abstract | Crossref Full Text | Google Scholar

134. Konijeti GG, Arora P, Boylan MR, Song Y, Huang S, Harrell F, et al. Vitamin D supplementation modulates T cell–mediated immunity in humans: results from a randomized control trial. J Clin Endocrinol Metab. (2016) 101:533–8. doi: 10.1210/jc.2015-3599

PubMed Abstract | Crossref Full Text | Google Scholar

135. Abdelhafez OH, Fahim JR, Mustafa M, AboulMagd AM, Desoukey SY, Hayallah AM, et al. Natural metabolites from the soft coral Nephthea sp as potential SARS-CoV-2 main protease inhibitors. Nat Prod Res. (2021) 36:2893–6. doi: 10.1080/14786419.2021.1925892

PubMed Abstract | Crossref Full Text | Google Scholar

136. El-Hawary SS, Issa MY, Ebrahim HS, Mohammed AF, Hayallah AM, El-Kadder EMA, et al. Potential of (Citrus nobilis Lour × Citrus deliciosa Tenora) metabolites on COVID-19 virus main protease supported by in silico analysis. Nat Prod Res. (2021) 36:2843–7. doi: 10.1080/14786419.2021.1917573

PubMed Abstract | Crossref Full Text | Google Scholar

137. Hassan HA, Abdelmohsen UR, Aly OM, Desoukey SY, Mohamed KM, Kamel MS. Potential of Ficus microcarpa metabolites against SARS-CoV-2 main protease supported by docking studies. Nat Prod Res. (2022) 36:994–8. doi: 10.1080/14786419.2020.1839452

PubMed Abstract | Crossref Full Text | Google Scholar

138. Sayed AM, Ibrahim AH, Tajuddeen N, Seibel J, Bodem J, Geiger N, et al. Korupensamine A, but not its atropisomer, korupensamine B, inhibits SARS-CoV-2 in vitro by targeting its main protease (Mpro). Eur J Med Chem. (2023) 251:115226. doi: 10.1016/j.ejmech.2023.115226

PubMed Abstract | Crossref Full Text | Google Scholar

139. Zahran EM, Sayed AM, Abdelwahab MF, Albohy A, Abdulrazik BS, Ibrahim AM, et al. Identifying the specific-targeted marine cerebrosides against SARS-CoV-2: an integrated computational approach. RSC Adv. (2021) 11:36042–59. doi: 10.1039/D1RA07103C

PubMed Abstract | Crossref Full Text | Google Scholar

140. Zahran EM, Albohy A, Khalil A, Ibrahim AH, Ahmed HA, El-Hossary EM, et al. Bioactivity potential of marine natural products from scleractinia-associated microbes and in silico anti-SARS-COV-2 evaluation. Mar Drugs. (2020) 18:645. doi: 10.3390/md18120645

PubMed Abstract | Crossref Full Text | Google Scholar

141. El-Hawary SS, Ali TFS, Abo El-Ela SO, Elwekeel A, Abdelmohsen UR, Owis AI. Secondary metabolites of Livistona decipiens as potential inhibitors of SARS-CoV-2. RSC Adv. (2022) 12:19505–11. doi: 10.1039/D2RA01306A

PubMed Abstract | Crossref Full Text | Google Scholar

142. ElNaggar MH, Abdelwahab GM, Kutkat O, GabAllah M, Ali MA, El-Metwally MEA, et al. Aurasperone A inhibits SARS CoV-2 in vitro: an integrated in vitro and in silico study. Mar Drugs. (2022) 20:179. doi: 10.3390/md20030179

PubMed Abstract | Crossref Full Text | Google Scholar

143. El-Hawary SS, Rabeh MA, Raey MAE, El-Kadder EMA, Sobeh M, Abdelmohsen UR, et al. Metabolomic profiling of three Araucaria species, and their possible potential role against COVID-19. J Biomol Struct Dyn. (2022) 40:6426–38. doi: 10.1080/07391102.2021.1885494

PubMed Abstract | Crossref Full Text | Google Scholar

144. Adel A, Elnaggar MS, Albohy A, Elrashedy AA, Mostafa A, Kutkat O, et al. Evaluation of antiviral activity of Carica papaya leaves against SARS-CoV-2 assisted by metabolomic profiling. RSC Adv. (2022) 12:32844–52. doi: 10.1039/D2RA04600H

PubMed Abstract | Crossref Full Text | Google Scholar

145. Albohy A, Zahran EM, Abdelmohsen UR, Salem MA, Al-Warhi T, Al-Sanea MM, et al. Multitarget in silico studies of Ocimum menthiifolium, family Lamiaceae against SARS-CoV-2 supported by molecular dynamics simulation. J Biomol Struct Dyn. (2022) 40:4062–72. doi: 10.1080/07391102.2020.1852964

PubMed Abstract | Crossref Full Text | Google Scholar

146. Al-Sanea MM, Abelyan N, Abdelgawad MA, Musa A, Ghoneim MM, Al-Warhi T, et al. Strawberry and ginger silver nanoparticles as potential inhibitors for SARS-CoV-2 assisted by in silico modeling and metabolic profiling. Antibiotics. (2021) 10:824. doi: 10.3390/antibiotics10070824

PubMed Abstract | Crossref Full Text | Google Scholar

147. Eltamany EE, Elhady SS, Goda MS, Aly OM, Habib ES, Ibrahim AK, et al. Chemical composition of the Red Sea Green Algae Ulva lactuca: isolation and in silico studies of new anti-COVID-19 ceramides. Metabolites. (2021) 11:816. doi: 10.3390/metabo11120816

PubMed Abstract | Crossref Full Text | Google Scholar

148. Shady NH, Hayallah AM, Mohamed MFA, Ghoneim MM, Chilingaryan G, Al-Sanea MM, et al. Targeting 3CLpro and SARS-CoV-2 RdRp by Amphimedon sp. metabolites: a computational study. Molecules. (2021) 26:3775. doi: 10.3390/molecules26123775

PubMed Abstract | Crossref Full Text | Google Scholar

149. Orfali R, Rateb ME, Hassan HM, Alonazi M, Gomaa MR, Mahrous N, et al. Sinapic acid suppresses SARS CoV-2 replication by targeting its envelope protein. Antibiotics. (2021) 10:420. doi: 10.3390/antibiotics10040420

PubMed Abstract | Crossref Full Text | Google Scholar

150. Sayed AM, Alhadrami HA, El-Gendy AO, Shamikh YI, Belbahri L, Hassan HM, et al. Microbial natural products as potential inhibitors of SARS-CoV-2 main protease (M(pro)). Microorganisms. (2020) 8:970. doi: 10.3390/microorganisms8070970

PubMed Abstract | Crossref Full Text | Google Scholar

151. Owis AI, El-Hawary MS, El Amir D, Aly OM, Abdelmohsen UR, Kamel MS. Molecular docking reveals the potential of Salvadora persica flavonoids to inhibit COVID-19 virus main protease. RSC Adv. (2020) 10:19570–5. doi: 10.1039/D0RA03582C

PubMed Abstract | Crossref Full Text | Google Scholar

152. El Hawary SS, Khattab AR, Marzouk HS, El Senousy AS, Alex MGA, Aly OM, et al. In silico identification of SARS-CoV-2 spike (S) protein-ACE2 complex inhibitors from eight Tecoma species and cultivars analyzed by LC-MS. RSC Adv. (2020) 10:43103–8. doi: 10.1039/D0RA08997D

PubMed Abstract | Crossref Full Text | Google Scholar

153. Abd El-Mordy FM, El-Hamouly MM, Ibrahim MT, El-Rheem GA, Aly OM, Abd El-Kader AM, et al. Inhibition of SARS-CoV-2 main protease by phenolic compounds from Manilkara hexandra (Roxb) Dubard assisted by metabolite profiling and in silico virtual screening. RSC Adv. (2020) 10:32148–55. doi: 10.1039/D0RA05679K

PubMed Abstract | Crossref Full Text | Google Scholar

154. Mullowney MW, Duncan KR, Elsayed SS, Garg N, van der Hooft JJJ, Martin NI, et al. Artificial intelligence for natural product drug discovery. Nat Rev Drug Discov. (2023) 22(11):895-916. doi: 10.1038/s41573-023-00774-7

PubMed Abstract | Crossref Full Text | Google Scholar

155. Yalçin S, Yalçinkaya S, Ercan F. Determination of potential drug candidate molecules of the Hypericum perforatum for COVID-19 treatment. Curr Pharmacol Rep. (2021) 7:42–8. doi: 10.1007/s40495-021-00254-9

PubMed Abstract | Crossref Full Text | Google Scholar

156. Samarth NR, Kabra S, Singh Anthraquinolone and quinolizine derivatives as an alley of future treatment for COVID-19: an in silico machine learning hypothesis. Sci Rep. (2021) 11:17915. doi: 10.1038/s41598-021-97031-x

PubMed Abstract | Crossref Full Text | Google Scholar

157. Banerjee S, Yadav S, Banerjee S, Fakayode SO, Parvathareddy J, Reichard W, et al. Drug repurposing to identify nilotinib as a potential SARS-CoV-2 main protease inhibitor: insights from a computational and in vitro study. J Chem Inform Model. (2021) 61:5469–83. doi: 10.1021/acs.jcim.1c00524

PubMed Abstract | Crossref Full Text | Google Scholar

158. Mandour YM, Zlotos DP, Alaraby Salem M, A. multi-stage virtual screening of FDA-approved drugs reveals potential inhibitors of SARS-CoV-2 main protease. J Biomol Struct Dyn. (2022) 40:2327–38. doi: 10.1080/07391102.2020.1837680

PubMed Abstract | Crossref Full Text | Google Scholar

159. Jo S, Kim S, Kim DY, Kim MS, Shin DH. Flavonoids with inhibitory activity against SARS-CoV-2 3CLpro. J Enzyme Inhib Med Chem. (2020) 35:1539–44. doi: 10.1080/14756366.2020.1801672

PubMed Abstract | Crossref Full Text | Google Scholar

160. Liu H, Ye F, Sun Q, Liang H, Li C, Li S, et al. Scutellaria baicalensis extract and baicalein inhibit replication of SARS-CoV-2 and its 3C-like protease in vitro. J Enzyme Inhib Med Chem. (2021) 36:497–503. doi: 10.1080/14756366.2021.1873977

PubMed Abstract | Crossref Full Text | Google Scholar

161. Chakraborty R, Bhattacharje G, Baral J, Manna B, Mullick J, Mathapati BS, et al. In-silico screening and in-vitro assay show the antiviral effect of Indomethacin against SARS-CoV-2. Comput Biol Med. (2022) 147:105788. doi: 10.1016/j.compbiomed.2022.105788

PubMed Abstract | Crossref Full Text | Google Scholar

162. Ibrahim MAA, Abdelrahman AHM, Atia MAM, Mohamed TA, Moustafa MF, Hakami AR, et al. Blue biotechnology: computational screening of sarcophyton cembranoid diterpenes for SARS-CoV-2 main protease inhibition. Mar Drugs. (2021) 19:391. doi: 10.3390/md19070391

PubMed Abstract | Crossref Full Text | Google Scholar

163. Tahir Ul Qamar M, Alqahtani SM, Alamri MA, Chen LL. Structural basis of SARS-CoV-2 3CLpro and anti-COVID-19 drug discovery from medicinal plants. J Pharm Anal. (2020) 10:313–9. doi: 10.1016/j.jpha.2020.03.009

PubMed Abstract | Crossref Full Text | Google Scholar

164. Vincent S, Arokiyaraj S, Saravanan M, Dhanraj M. Molecular docking studies on the anti-viral effects of compounds from kabasura kudineer on SARS-CoV-2 3CLpro. Front Mol Biosci. (2020) 7:613401. doi: 10.3389/fmolb.2020.613401

PubMed Abstract | Crossref Full Text | Google Scholar

165. Bharadwaj S, Dubey A, Yadava U, Mishra SK, Kang SG, Dwivedi VD. Exploration of natural compounds with anti-SARS-CoV-2 activity via inhibition of SARS-CoV-2 Mpro. Brief Bioinform. (2021) 22:1361–77. doi: 10.1093/bib/bbaa382

PubMed Abstract | Crossref Full Text | Google Scholar

166. Sharma A, Goyal S, Yadav AK, Kumar P, Gupta L. In-silico screening of plant-derived antivirals against main protease, 3CLpro and endoribonuclease, NSP15 proteins of SARS-CoV-2. J Biomol Struct Dyn. (2022) 40:86–100. doi: 10.1080/07391102.2020.1808077

PubMed Abstract | Crossref Full Text | Google Scholar

167. Nallusamy S, Mannu J, Ravikumar C, Angamuthu K, Nathan B, Nachimuthu K, et al. Exploring phytochemicals of traditional medicinal plants exhibiting inhibitory activity against main protease, spike glycoprotein, RNA-dependent RNA polymerase and non-structural proteins of SARS-CoV-2 through virtual screening. Front Pharmacol. (2021) 12:667704. doi: 10.3389/fphar.2021.667704

PubMed Abstract | Crossref Full Text | Google Scholar

168. Alhadrami HA, Sayed AM, Hassan HM, Youssif KA, Gaber Y, Moatasim Y, et al. Cnicin as an anti-SARS-CoV-2: an Integrated in silico and in vitro approach for the rapid identification of potential COVID-19 therapeutics. Antibiotics. (2021) 10:542. doi: 10.3390/antibiotics10050542

PubMed Abstract | Crossref Full Text | Google Scholar

169. Sinha SK, Prasad SK, Islam MA, Gurav SS, Patil RB, AlFaris NA, et al. Identification of bioactive compounds from Glycyrrhiza glabra as possible inhibitor of SARS-CoV-2 spike glycoprotein and non-structural protein-15: a pharmacoinformatics study. J Biomol Struct Dyn. (2021) 39:4686–700. doi: 10.1080/07391102.2020.1779132

PubMed Abstract | Crossref Full Text | Google Scholar

170. Abdelmohsen UR, Albohy A, Abdulrazik BS, Bayoumi SAL, Malak LG, Khallaf ISA, et al. Natural coumarins as potential anti-SARS-CoV-2 agents supported by docking analysis. RSC Adv. (2021) 11:16970–9. doi: 10.1039/D1RA01989A

PubMed Abstract | Crossref Full Text | Google Scholar

171. Chandra A, Chaudhary M, Qamar I, Singh N, Nain V. In silico identification and validation of natural antiviral compounds as potential inhibitors of SARS-CoV-2 methyltransferase. J Biomol Struct Dyn. (2022) 40:6534–44. doi: 10.1080/07391102.2021.1886174

PubMed Abstract | Crossref Full Text | Google Scholar

172. M P, Reddy GJ, Hema K, Dodoala S, Koganti B. Unravelling high-affinity binding compounds towards transmembrane protease serine 2 enzyme in treating SARS-CoV-2 infection using molecular modelling and docking studies. Eur J Pharmacol. (2021) 890:173688. doi: 10.1016/j.ejphar.2020.173688

PubMed Abstract | Crossref Full Text | Google Scholar

173. Dhanjal JK, Kumar V, Garg S, Subramani C, Agarwal S, Wang J, et al. Molecular mechanism of anti-SARS-CoV2 activity of Ashwagandha-derived withanolides. Int J Biol Macromol. (2021) 184:297–312. doi: 10.1016/j.ijbiomac.2021.06.015

PubMed Abstract | Crossref Full Text | Google Scholar

174. Huang X, Pearce R, Omenn GS, Zhang Y. Identification of 13 guanidinobenzoyl- or aminidinobenzoyl-containing drugs to potentially inhibit TMPRSS2 for COVID-19 treatment. Int J Mol Sci. (2021) 22:7060. doi: 10.3390/ijms22137060

PubMed Abstract | Crossref Full Text | Google Scholar

175. Malik A, Kohli M, Jacob NA, Kayal A, Raj TK, Kulkarni N, et al. In silico screening of phytochemical compounds and FDA drugs as potential inhibitors for NSP16/10 5' methyl transferase activity. J Biomol Struct Dyn. (2023) 41:221–33. doi: 10.1080/07391102.2021.2005680

PubMed Abstract | Crossref Full Text | Google Scholar

176. Yi Y, Zhang M, Xue H, Yu R, Bao YO, Kuang Y, et al. Schaftoside inhibits 3CLpro and PLpro of SARS-CoV-2 virus and regulates immune response and inflammation of host cells for the treatment of COVID-19. Acta Pharm Sin B. (2022) 12:4154–64. doi: 10.1016/j.apsb.2022.07.017

PubMed Abstract | Crossref Full Text | Google Scholar

177. Hisham Shady N, Youssif KA, Sayed AM, Belbahri L, Oszako T, Hassan HM, et al. Sterols and triterpenes: antiviral potential supported by in-silico analysis. Plants. (2021) 10:41. doi: 10.3390/plants10010041

PubMed Abstract | Crossref Full Text | Google Scholar

178. Gao H, Dai R, Su R. Computer-aided drug design for the pain-like protease (PLpro) inhibitors against SARS-CoV-2. Biomed Pharmacother. (2023) 159:114247. doi: 10.1016/j.biopha.2023.114247

PubMed Abstract | Crossref Full Text | Google Scholar

179. Rivero-Segura NA, Gomez-Verjan JC. In silico screening of natural products isolated from Mexican herbal medicines against COVID-19. Biomolecules. (2021) 11:216. doi: 10.3390/biom11020216

PubMed Abstract | Crossref Full Text | Google Scholar

180. Gogoi M, Borkotoky M, Borchetia S, Chowdhury P, Mahanta S, Barooah AK. Black tea bioactives as inhibitors of multiple targets of SARS-CoV-2 (3CLpro, PLpro and RdRp): a virtual screening and molecular dynamic simulation study. J Biomol Struct Dyn. (2022) 40:7143–66. doi: 10.1080/07391102.2021.1897679

PubMed Abstract | Crossref Full Text | Google Scholar

181. Sharma A, Vora J, Patel D, Sinha S, Jha PC, Shrivastava N. Identification of natural inhibitors against prime targets of SARS-CoV-2 using molecular docking, molecular dynamics simulation and MM-PBSA approaches. J Biomol Struct Dyn. (2022) 40:3296–311. doi: 10.1080/07391102.2020.1846624

PubMed Abstract | Crossref Full Text | Google Scholar

182. Joshi RS, Jagdale S, Bansode S, Shankar SS, Tellis M, Pandya VK, et al. Discovery of potential multi-target-directed ligands by targeting host-specific SARS-CoV-2 structurally conserved main protease. J Biomol Struct Dyn. (2021) 39:3099–114. doi: 10.20944/preprints202004.0068.v2

PubMed Abstract | Crossref Full Text | Google Scholar

183. Emirik M. Potential therapeutic effect of turmeric contents against SARS-CoV-2 compared with experimental COVID-19 therapies: in silico study. J Biomol Struct Dyn. (2022) 40:2024–37. doi: 10.1080/07391102.2020.1835719

PubMed Abstract | Crossref Full Text | Google Scholar

184. Latha D, Hrishikesh D, Shiban G, Chandrashekar C, Bharath BR. In silico, in vitro screening of plant extracts for anti-SARS-CoV-2 activity and evaluation of their acute and sub-acute toxicity. Phytomed Plus. (2022) 2:100233. doi: 10.1016/j.phyplu.2022.100233

PubMed Abstract | Crossref Full Text | Google Scholar

185. Hamdy R, Mostafa A, Abo Shama NM, Soliman SSM, Fayed B. Comparative evaluation of flavonoids reveals the superiority and promising inhibition activity of silibinin against SARS-CoV-2. Phytother Res. (2022) 36:2921–39. doi: 10.1002/ptr.7486

PubMed Abstract | Crossref Full Text | Google Scholar

186. Yepes-Pérez AF, Herrera-Calderon O, Quintero-Saumeth J. Uncaria tomentosa (cat's claw): a promising herbal medicine against SARS-CoV-2/ACE-2 junction and SARS-CoV-2 spike protein based on molecular modeling. J Biomol Struct Dyn. (2022) 40:2227–43. doi: 10.1080/07391102.2020.1837676

PubMed Abstract | Crossref Full Text | Google Scholar

187. Rajaraman D, Anthony LA, Nethaji P, Vallangi R. One-pot synthesis, NMR, quantum chemical approach, molecular docking studies, drug-likeness and in-silico ADMET prediction of novel 1-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)-2-(furan-2-yl)-4,5-diphenyl-1H-imidazole derivatives. J Mol Struct. (2023) 1273:134314. doi: 10.1016/j.molstruc.2022.134314

PubMed Abstract | Crossref Full Text | Google Scholar

188. Sabzian-Molaei F, Hosseini S, Alipour A, Ghaderi H, Fotouhi-Chahouki F, Hadi A, et al. Urtica dioica agglutinin (UDA) as a potential candidate for inhibition of SARS-CoV-2 Omicron variants: in silico prediction and experimental validation. Phytomedicine. (2023) 111:154648. doi: 10.1016/j.phymed.2023.154648

PubMed Abstract | Crossref Full Text | Google Scholar

189. Qiao J, Li YS, Zeng R, Liu FL, Luo RH, Huang C, et al. SARS-CoV-2 M(pro) inhibitors with antiviral activity in a transgenic mouse model. Science. (2021) 371:1374–8. doi: 10.1126/science.abf1611

PubMed Abstract | Crossref Full Text | Google Scholar

190. Fu L, Ye F, Feng Y, Yu F, Wang Q, Wu Y, et al. Both Boceprevir and GC376 efficaciously inhibit SARS-CoV-2 by targeting its main protease. Nat Commun. (2020) 11:4417. doi: 10.1038/s41467-020-18233-x

PubMed Abstract | Crossref Full Text | Google Scholar

191. Ongtanasup T, Wanmasae S, Srisang S, Manaspon C, Net-Anong S, Eawsakul K. In silico investigation of ACE2 and the main protease of SARS-CoV-2 with phytochemicals from Myristica fragrans (Houtt) for the discovery of a novel COVID-19 drug Saudi. J Biol Sci. (2022) 29:103389. doi: 10.1016/j.sjbs.2022.103389

PubMed Abstract | Crossref Full Text | Google Scholar

192. Tito A, Colantuono A, Pirone L, Pedone E, Intartaglia D, Giamundo G, et al. Pomegranate peel extract as an inhibitor of SARS-CoV-2 spike binding to human ACE2 receptor (in vitro): a promising source of novel antiviral drugs. Front Chem. (2021) 9:638187. doi: 10.3389/fchem.2021.638187

PubMed Abstract | Crossref Full Text | Google Scholar

193. Gangadevi S, Badavath VN, Thakur A, Yin N, De Jonghe S, Acevedo O, et al. Kobophenol A inhibits binding of host ACE2 receptor with spike RBD domain of SARS-CoV-2, a lead compound for blocking COVID-19. J Phys Chem Lett. (2021) 12:1793–802. doi: 10.1021/acs.jpclett.0c03119

PubMed Abstract | Crossref Full Text | Google Scholar

194. Thoene J, Gavin RF, Towne A, Wattay L, Ferrari MG, Navarrete J, et al. In vitro activity of cysteamine against SARS-CoV-2 variants. Mol Genet Metab. (2022) 137:192–200. doi: 10.1016/j.ymgme.2022.08.009

PubMed Abstract | Crossref Full Text | Google Scholar

195. Lim CT, Tan KW, Wu M, Ulferts R, Armstrong LA, Ozono E, et al. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of Nsp3 papain-like protease. Biochem J. (2021) 478:2517–31. doi: 10.1042/BCJ20210244

PubMed Abstract | Crossref Full Text | Google Scholar

196. Zhao Y, Du X, Duan Y, Pan X, Sun Y, You T, et al. High-throughput screening identifies established drugs as SARS-CoV-2 PLpro inhibitors. Protein Cell. (2021) 12:877–88. doi: 10.1007/s13238-021-00836-9

PubMed Abstract | Crossref Full Text | Google Scholar

197. Malebari AM, E A Ahmed H, Ihmaid SK, Omar AM, Muhammad YA, Althagfan SS, et al. Exploring the dual effect of novel 1,4-diarylpyranopyrazoles as antiviral and anti-inflammatory for the management of SARS-CoV-2 and associated inflammatory symptoms. Bioorg Chem. (2023) 130:106255. doi: 10.1016/j.bioorg.2022.106255

PubMed Abstract | Crossref Full Text | Google Scholar

198. Shang W, Dai W, Yao C, Xu L, Tao X, Su H, et al. In vitro and in vivo evaluation of the main protease inhibitor FB2001 against SARS-CoV-2. Antiviral Res. (2022) 208:105450. doi: 10.1016/j.antiviral.2022.105450

PubMed Abstract | Crossref Full Text | Google Scholar

199. Owen DR, Allerton CMN, Andersson AS, Aschenbrenner L, Avery M, Berritt S, et al. An oral SARS-CoV-2 M(pro) inhibitor clinical candidate for the treatment of COVID-19. Science. (2021) 374:1586–93. doi: 10.1126/science.abl4784

PubMed Abstract | Crossref Full Text | Google Scholar

200. Abdelnabi R, Foo CS, Jochmans D, Vangeel L, De Jonghe S, Augustijns P, et al. The oral protease inhibitor (PF-07321332) protects Syrian hamsters against infection with SARS-CoV-2 variants of concern. Nat Commun. (2022) 13:719. doi: 10.1038/s41467-022-28354-0

PubMed Abstract | Crossref Full Text | Google Scholar

201. Sasaki M, Tabata K, Kishimoto M, Itakura Y, Kobayashi H, Ariizumi T, et al. S-217622, a SARS-CoV-2 main protease inhibitor, decreases viral load and ameliorates COVID-19 severity in hamsters. Sci Transl Med. (2023) 15:eabq4064. doi: 10.1126/scitranslmed.abq4064

PubMed Abstract | Crossref Full Text | Google Scholar

202. Wang G, Yang ML, Duan ZL, Liu FL, Jin L, Long CB, et al. Dalbavancin binds ACE2 to block its interaction with SARS-CoV-2 spike protein and is effective in inhibiting SARS-CoV-2 infection in animal models. Cell Res. (2021) 31:17–24. doi: 10.1038/s41422-020-00450-0

PubMed Abstract | Crossref Full Text | Google Scholar

203. Arya R, Prashar V, Kumar M. Identification and characterization of aurintricarboxylic acid as a potential inhibitor of SARS-CoV-2 PLpro. Int J Biol Macromol. (2023) 2023:123347. doi: 10.1016/j.ijbiomac.2023.123347

PubMed Abstract | Crossref Full Text | Google Scholar

204. Schäfer A, Martinez DR, Won JJ, Meganck RM, Moreira FR, Brown AJ, et al. Therapeutic treatment with an oral prodrug of the remdesivir parental nucleoside is protective against SARS-CoV-2 pathogenesis in mice. Sci Transl Med. (2022) 14:eabm3410. doi: 10.1126/scitranslmed.abm3410

PubMed Abstract | Crossref Full Text | Google Scholar

205. Wahl A, Gralinski LE, Johnson CE, Yao W, Kovarova M, Dinnon KH, et al. SARS-CoV-2 infection is effectively treated and prevented by EIDD-2801. Nature. (2021) 591:451–7. doi: 10.1038/s41586-021-03312-w

PubMed Abstract | Crossref Full Text | Google Scholar

206. Johnson DM, Brasel T, Massey S, Garron T, Grimes M, Smith J, et al. Evaluation of molnupiravir (EIDD-2801) efficacy against SARS-CoV-2 in the rhesus macaque model. Antiviral Res. (2023) 209:105492. doi: 10.1016/j.antiviral.2022.105492

PubMed Abstract | Crossref Full Text | Google Scholar

207. Chan JF, Oh YJ, Yuan S, Chu H, Yeung ML, Canena D, et al. A molecularly engineered, broad-spectrum anti-coronavirus lectin inhibits SARS-CoV-2 and MERS-CoV infection in vivo. Cell Rep Med. (2022) 3:100774. doi: 10.1016/j.xcrm.2022.100774

PubMed Abstract | Crossref Full Text | Google Scholar

208. National Institute of Allergy and Infectious Diseases. Decades in the Making: mRNA COVID-19 Vaccines (2023) Available online at: https://www.niaid.nih.gov/diseases-conditions/decades-making-mrna-covid-19-vaccines

Google Scholar

209. Ball P. The lightning-fast quest for COVID vaccines — and what it means for other diseases. Nature. (2020) 589:16–8. doi: 10.1038/d41586-020-03626-1

PubMed Abstract | Crossref Full Text | Google Scholar

210. U.S. Food and Drug Administration. FDA Approves First COVID-19 Vaccine (2021). Available online at: https://www.fda.gov/news-events/press-announcements/fda-approves-first-covid-19-vaccine#:~:text=Since%20Dec.age%20on%20May%2010%2C%202021

Google Scholar

211. Apio C, Han K, Heo G, Park T. A statistical look at the COVID-19 vaccine development and vaccine policies. Front Public Health. (2022) 10:1048062. doi: 10.3389/fpubh.2022.1048062

PubMed Abstract | Crossref Full Text | Google Scholar

212. Allahyari F, Molaee H, Hosseini Nejad J. COVID-19 vaccines and neurological complications: a systematic review. Z Naturforsch C J Biosci. (2023) 78:1–8. doi: 10.1515/znc-2022-0092

PubMed Abstract | Crossref Full Text | Google Scholar

213. Lamprinou M, Sachinidis A, Stamoula E, Vavilis T, Papazisis G. COVID-19 vaccines adverse events: potential molecular mechanisms. Immunol Res. (2023) 71:356–72. doi: 10.1007/s12026-023-09357-5

PubMed Abstract | Crossref Full Text | Google Scholar

214. Fang E, Liu X, Li M, Zhang Z, Song L, Zhu B, et al. Advances in COVID-19 mRNA vaccine development. Signal Transduct Target Ther. (2022) 7:94. doi: 10.1038/s41392-022-00950-y

PubMed Abstract | Crossref Full Text | Google Scholar

215. Le T, Sun C, Chang J, Zhang G, Yin X. mRNA vaccine development for emerging animal and zoonotic diseases. Viruses. (2022) 14:401. doi: 10.3390/v14020401

PubMed Abstract | Crossref Full Text | Google Scholar

216. Park JW, Lagniton PNP, Liu Y, Xu RH. mRNA vaccines for COVID-19: what, why and how. Int J Biol Sci. (2021) 17:1446–60. doi: 10.7150/ijbs.59233

PubMed Abstract | Crossref Full Text | Google Scholar

217. Szabó GT, Mahiny AJ, Vlatkovic I. COVID-19 mRNA vaccines: platforms and current developments. Mol Ther. (2022) 30:1850–68. doi: 10.1016/j.ymthe.2022.02.016

PubMed Abstract | Crossref Full Text | Google Scholar

218. Huang Q, Zeng J, Yan J. COVID-19 mRNA vaccines. J Genet Genomics. (2021) 48:107–14. doi: 10.1016/j.jgg.2021.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

219. Qu L, Yi Z, Shen Y, Lin L, Chen F, Xu Y, et al. Circular RNA vaccines against SARS-CoV-2 and emerging variants. Cell. (2022) 185:1728–44.e16. doi: 10.1016/j.cell.2022.03.044

PubMed Abstract | Crossref Full Text | Google Scholar

220. Liu X, Zhang Y, Zhou S, Dain L, Mei L, Zhu G. Circular RNA: an emerging frontier in RNA therapeutic targets, RNA therapeutics, and mRNA vaccines. J Control Release. (2022) 348:84–94. doi: 10.1016/j.jconrel.2022.05.043

PubMed Abstract | Crossref Full Text | Google Scholar

221. Zhao X, Zhong Y, Wang X, Shen J, An W. Advances in circular RNA and its applications. Int J Med Sci. (2022) 19:975–85. doi: 10.7150/ijms.71840

PubMed Abstract | Crossref Full Text | Google Scholar

222. Bai Y, Liu D, He Q, Liu J, Mao Q, Liang Z. Research progress on circular RNA vaccines. Front Immunol. (2022) 13:1091797. doi: 10.3389/fimmu.2022.1091797

PubMed Abstract | Crossref Full Text | Google Scholar

223. Breuer J, Barth P, Noe Y, Shalamova L, Goesmann A, Weber F, et al. What goes around comes around: artificial circular RNAs bypass cellular antiviral responses. Mol Ther Nucleic Acids. (2022) 28:623–35. doi: 10.1016/j.omtn.2022.04.017

PubMed Abstract | Crossref Full Text | Google Scholar

224. Bhattacharya M, Sharma AR, Ghosh P, Patra P, Patra BC, Lee S, et al. Bioengineering of novel non-replicating mRNA (NRM) and self-amplifying mRNA (SAM) vaccine candidates against SARS-CoV-2 using immunoinformatics approach. Mol Biotechnol. (2022) 64:510–25. doi: 10.1007/s12033-021-00432-6

PubMed Abstract | Crossref Full Text | Google Scholar

225. Guo X, Liu D, Huang Y, Deng Y, Wang Y, Mao J, et al. Revolutionizing viral disease vaccination: the promising clinical advancements of non-replicating mRNA vaccines. Virol J. (2023) 20:64. doi: 10.1186/s12985-023-02023-0

PubMed Abstract | Crossref Full Text | Google Scholar

226. Blakney AK, Ip S, Geall AJ. An update on self-amplifying mRNA vaccine development. Vaccines. (2021) 9:97. doi: 10.3390/vaccines9020097

PubMed Abstract | Crossref Full Text | Google Scholar

227. Pourseif MM, Masoudi-Sobhanzadeh Y, Azari E, Parvizpour S, Barar J, Ansari R, et al. Self-amplifying mRNA vaccines: mode of action, design, development and optimization. Drug Discov Today. (2022) 27:103341. doi: 10.1016/j.drudis.2022.103341

PubMed Abstract | Crossref Full Text | Google Scholar

228. Bloom K, van den Berg F, Arbuthnot P. Self-amplifying RNA vaccines for infectious diseases. Gene Ther. (2021) 28:117–29. doi: 10.1038/s41434-020-00204-y

PubMed Abstract | Crossref Full Text | Google Scholar

229. Coughlan L. Factors which contribute to the immunogenicity of non-replicating adenoviral vectored vaccines. Front Immunol. (2020) 11:909. doi: 10.3389/fimmu.2020.00909

PubMed Abstract | Crossref Full Text | Google Scholar

230. Kulanayake S, Tikoo SK. Adenovirus core proteins: structure and function. Viruses. (2021) 13:338. doi: 10.3390/v13030388

PubMed Abstract | Crossref Full Text | Google Scholar

231. Sakurai F, Tachibana M, Mizuguchi H. Adenovirus vector-based vaccine for infectious diseases. Drug Metab Pharmacokinet. (2022) 42:100432. doi: 10.1016/j.dmpk.2021.100432

PubMed Abstract | Crossref Full Text | Google Scholar

232. Jacob-Dolan C, Barouch DH. COVID-19 vaccines: adenoviral vectors. Annu Rev Med. (2022) 73:41–54. doi: 10.1146/annurev-med-012621-102252

PubMed Abstract | Crossref Full Text | Google Scholar

233. Zhong J, Liu S, Cui T, Li J, Zhu F, Zhong N, et al. Heterologous booster with inhaled adenovirus vector COVID-19 vaccine generated more neutralizing antibodies against different SARS-CoV-2 variants. Emerg Microbes Infect. (2022) 11:2689–97. doi: 10.1080/22221751.2022.2132881

PubMed Abstract | Crossref Full Text | Google Scholar

234. Hotez PJ, Bottazzi ME. Whole inactivated virus and protein-based COVID-19 vaccines. Annu Rev Med. (2022) 73:55–64. doi: 10.1146/annurev-med-042420-113212

PubMed Abstract | Crossref Full Text | Google Scholar

235. Li M, Wang H, Tian L, Pang Z, Yang Q, Huang T, et al. COVID-19 vaccine development: milestones, lessons and prospects. Signal Transduct Target Ther. (2022) 7:146. doi: 10.1038/s41392-022-00996-y

PubMed Abstract | Crossref Full Text | Google Scholar

236. Hadj Hassine I. COVID-19 vaccines and variants of concern: a review. Rev Med Virol. (2022) 32:e2313. doi: 10.1002/rmv.2313

PubMed Abstract | Crossref Full Text | Google Scholar

237. Heidary M, Kaviar VH, Shirani M, Ghanavati R, Motahar M, Sholeh M, et al. A comprehensive review of the protein subunit vaccines against COVID-19. Front Microbiol. (2022) 13:927306. doi: 10.3389/fmicb.2022.927306

PubMed Abstract | Crossref Full Text | Google Scholar

238. Sandoval C, Guerrero D, Muñoz J, Godoy K, Souza-Mello V, Farías J. Effectiveness of mRNA, protein subunit vaccine and viral vectors vaccines against SARS-CoV-2 in people over 18 years old: a systematic review. Expert Rev Vaccines. (2023) 22:35–53. doi: 10.1080/14760584.2023.2156861

PubMed Abstract | Crossref Full Text | Google Scholar

239. Dar HA, Waheed Y, Najmi MH, Ismail S, Hetta HF, Ali A, et al. Multiepitope Subunit Vaccine Design against COVID-19 Based on the Spike Protein of SARS-CoV-2: an. J Immunol Res. (2020) 2020:8893483. doi: 10.1155/2020/8893483

PubMed Abstract | Crossref Full Text | Google Scholar

240. Silveira MM, Moreira GMSG, Mendonça M, DNA. vaccines against COVID-19: perspectives and challenges. Life Sci. (2021) 267:118919. doi: 10.1016/j.lfs.2020.118919

PubMed Abstract | Crossref Full Text | Google Scholar

241. Aurisicchio L, Brambilla N, Cazzaniga ME, Bonfanti P, Milleri S, Ascierto PA, et al. A first-in-human trial on the safety and immunogenicity of COVID-eVax, a cellular response-skewed DNA vaccine against COVID-19. Mol Ther. (2023) 31:788–800. doi: 10.1016/j.ymthe.2022.12.017

PubMed Abstract | Crossref Full Text | Google Scholar

242. Gatechompol S, Kittanamongkolchai W, Ketloy C, Prompetchara E, Thitithanyanont A, Jongkaewwattana A, et al. Safety and immunogenicity of a prefusion non-stabilized spike protein mRNA COVID-19 vaccine: a phase I trial. Nat Microbiol. (2022) 7:1987–95. doi: 10.1038/s41564-022-01271-0

PubMed Abstract | Crossref Full Text | Google Scholar

243. Joshi G Borah P Thakur S Sharma P Mayank andPoduri R Exploring the COVID-19 vaccine candidates against SARS-CoV-2 and its variants: where do we stand and where do we go? Hum Vaccin Immunother. (2021) 17:4714–40. doi: 10.1080/21645515.2021.1995283

PubMed Abstract | Crossref Full Text | Google Scholar

244. World Health Organization. COVID-19 Vaccine Tracker and Landscape (2023). Available online at: https://www.who.int/publications/m/item/draft-landscape-of-covid-19-candidate-vaccines

Google Scholar

245. Centers for Disease Control and Prevention. Overview of COVID-19 Vaccines (2023). Available online at: https://www.cdc.gov/covid/hcp/vaccine-considerations/overview.html

Google Scholar

246. Rashedi R, Samieefar N, Masoumi N, Mohseni S, Rezaei N. COVID-19 vaccines mix-and-match: the concept, the efficacy and the doubts. J Med Virol. (2022) 94:1294–9. doi: 10.1002/jmv.27463

PubMed Abstract | Crossref Full Text | Google Scholar

247. Centers for Disease Control and Prevention. Safety of COVID-19 Vaccines (2023). Available online at: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/safety/safety-of-vaccines.html

Google Scholar

248. Centers for Disease Control and Prevention. Possible Side Effects After Getting a COVID-19 Vaccine (2023). Available online at https://www.cdc.gov/coronavirus/2019-ncov/vaccines/expect/after.html

Google Scholar

249. Centers for Disease Control and Prevention. Selected Adverse Events Reported after COVID-19 Vaccination (2023). Available online at: https://www.cdc.gov/coronavirus/2019-ncov/vaccines/safety/adverse-events.html

Google Scholar

250. Piccialli F, di Cola VS, Giampaolo F, Cuomo S. The role of artificial intelligence in fighting the COVID-19 pandemic information systems. Frontiers. (2021) 23:1467–97. doi: 10.1007/s10796-021-10131-x

PubMed Abstract | Crossref Full Text | Google Scholar

251. Naseem M, Akhund R, Arshad H, Ibrahim MT. Exploring the potential of artificial intelligence and machine learning to combat COVID-19 and existing opportunities for LMIC: a scoping review. J Prim Care Commun Health. (2020) 11:2150132720963634. doi: 10.1177/2150132720963634

PubMed Abstract | Crossref Full Text | Google Scholar

252. Waheed A, Shafi J. Successful role of smart technology to combat COVID-19. In: 2020 fourth International Conference on I-SMAC (IoT in Social, Mobile, Analytics and Cloud)(I-SMAC). Palladam: IEEE (2020). doi: 10.1109/I-SMAC49090.2020.9243444

Crossref Full Text | Google Scholar

253. Kimura Y, Seki T, Miyata S, Arai Y, Murata T, Inoue H, et al. Hotspot analysis of COVID-19 infection using mobile-phone location data. Artif Life Robot. (2023) 28:43–9. doi: 10.1007/s10015-022-00830-2

PubMed Abstract | Crossref Full Text | Google Scholar

254. Feng C, Wang L, Chen X, Zhai Y, Zhu F, Chen H, et al. A novel artificial intelligence-assisted triage tool to aid in the diagnosis of suspected COVID-19 pneumonia cases in fever clinics. Ann Transl Med. (2021) 9:201. doi: 10.21037/atm-20-3073

PubMed Abstract | Crossref Full Text | Google Scholar

255. Abbasi Habashi S, Koyuncu M, Alizadehsani R. A survey of COVID-19 diagnosis using routine blood tests with the aid of artificial intelligence techniques. Diagnostics. (2023) 13:1749. doi: 10.3390/diagnostics13101749

PubMed Abstract | Crossref Full Text | Google Scholar

256. Shi F, Wang J, Shi J, Wu Z, Wang Q, Tang Z, et al. Review of artificial intelligence techniques in imaging data acquisition, segmentation, and diagnosis for COVID-19. IEEE Rev Biomed Eng. (2021) 14:4–15. doi: 10.1109/RBME.2020.2987975

PubMed Abstract | Crossref Full Text | Google Scholar

257. Alharthi, H. Predicting the level of generalized anxiety disorder of the coronavirus pandemic among college age students using artificial intelligence technology. In: 2020 19th International Symposium on Distributed Computing and Applications for Business Engineering and Science (DCABES). Xuzhou: IEEE (2020). doi: 10.1109/DCABES50732.2020.00064

Crossref Full Text | Google Scholar

258. Sau A, Bhakta I. Screening of anxiety and depression among seafarers using machine learning technology. Inform Med Unlocked. (2019) 16:100228. doi: 10.1016/j.imu.2019.100228

Crossref Full Text | Google Scholar

259. Ortiz-Barrios M, Arias-Fonseca S, Ishizaka A, Barbati M, Avendaño-Collante B, Navarro-Jiménez E. Artificial intelligence and discrete-event simulation for capacity management of intensive care units during the COVID-19 pandemic: a case study. J Business Res. (2023) 160:113806. doi: 10.1016/j.jbusres.2023.113806

PubMed Abstract | Crossref Full Text | Google Scholar

260. Annamalai A, Karuppaiya V, Ezhumalai D, Cheruparambath P, Balakrishnan K, Venkatesan A. Nano-based techniques: a revolutionary approach to prevent COVID-19 and enhancing human awareness. J Drug Deliv Sci Technol. (2023) 86:104567. doi: 10.1016/j.jddst.2023.104567

PubMed Abstract | Crossref Full Text | Google Scholar

261. Tang Z, Zhang X, Shu Y, Guo M, Zhang H, Tao W. Insights from nanotechnology in COVID-19 treatment. Nano Today. (2021) 36:101019. doi: 10.1016/j.nantod.2020.101019

PubMed Abstract | Crossref Full Text | Google Scholar

262. Hossain MK, Khan MI, El-Denglawey A, A. review on biomedical applications, prospects, and challenges of rare earth oxides. Appl Mater Today. (2021) 24:101104. doi: 10.1016/j.apmt.2021.101104

Crossref Full Text | Google Scholar

263. Anik MI, Mahmud N, Masud AA, Hasan M. Gold nanoparticles (GNPs) in biomedical and clinical applications: a review. Nano Select. (2022) 3:792–828. doi: 10.1002/nano.202100255

Crossref Full Text | Google Scholar

264. Lew TTS, Aung KMM, Ow SY, Amrun SN, Sutarlie L, Ng LFP, et al. Epitope-functionalized gold nanoparticles for rapid and selective detection of SARS-CoV-2 IgG antibodies. ACS Nano. (2021) 15:12286–97. doi: 10.1021/acsnano.1c04091

PubMed Abstract | Crossref Full Text | Google Scholar

265. Pramanik A, Gao Y, Patibandla S, Mitra D, McCandless MG, Fassero LA, et al. The rapid diagnosis and effective inhibition of coronavirus using spike antibody attached gold nanoparticles. Nanoscale Adv. (2021) 3:1588–96. doi: 10.1039/D0NA01007C

PubMed Abstract | Crossref Full Text | Google Scholar

266. Somvanshi SB, Kharat PB, Saraf TS, Jadhav KM, Somwanshi SB, Shejul SB. Multifunctional nano-magnetic particles assisted viral RNA-extraction protocol for potential detection of COVID-19. Mater Res Innov. (2021) 25:169–74. doi: 10.1080/14328917.2020.1769350

Crossref Full Text | Google Scholar

267. Wang Z, Zheng Z, Hu H, Zhou Q, Liu W, Li X, et al. A point-of-care selenium nanoparticle-based test for the combined detection of anti-SARS-CoV-2 IgM and IgG in human serum and blood. Lab Chip. (2020) 20:4255–61. doi: 10.1039/D0LC00828A

PubMed Abstract | Crossref Full Text | Google Scholar

268. Patel S, Srivastav AK, Gupta SK, Kumar U, Mahapatra SK, Gajjar PN, et al. Carbon nanotubes for rapid capturing of SARS-COV-2 virus: revealing a mechanistic aspect of binding based on computational studies. RSC Adv. (2021) 11:5785–800. doi: 10.1039/D0RA08888A

PubMed Abstract | Crossref Full Text | Google Scholar

269. Zamzami MA, Rabbani G, Ahmad A, Basalah AA, Al-Sabban WH, Nate Ahn S, et al. Carbon nanotube field-effect transistor (CNT-FET)-based biosensor for rapid detection of SARS-CoV-2 (COVID-19) surface spike protein S1. Bioelectrochemistry. (2022) 143:107982. doi: 10.1016/j.bioelechem.2021.107982

PubMed Abstract | Crossref Full Text | Google Scholar

270. Drobysh M, Ramanaviciene A, Viter R, Ramanavicius A. Affinity sensors for the diagnosis of COVID-19. Micromachines. (2021) 12:390. doi: 10.3390/mi12040390

PubMed Abstract | Crossref Full Text | Google Scholar

271. La Marca A, Capuzzo M, Paglia T, Roli L, Trenti T, Nelson SM. Testing for SARS-CoV-2 (COVID-19): a systematic review and clinical guide to molecular and serological in-vitro diagnostic assays. Reprod BioMed. (2020) 41:483–99. doi: 10.1016/j.rbmo.2020.06.001

PubMed Abstract | Crossref Full Text | Google Scholar

272. Murugan D, Bhatia H, Sai VVR, Satija J. P-FAB: a fiber-optic biosensor device for rapid detection of COVID-19. Trans Indian Natl Acad Eng. (2020) 5:211–5. doi: 10.1007/s41403-020-00122-w

PubMed Abstract | Crossref Full Text | Google Scholar

273. Tsakiri M, Naziris N, Demetzos C. Innovative vaccine platforms against infectious diseases: under the scope of the COVID-19 pandemic. Int J Pharm. (2021) 610:121212. doi: 10.1016/j.ijpharm.2021.121212

PubMed Abstract | Crossref Full Text | Google Scholar

274. Diaz-Arévalo, D. and M. Zeng, Chapter 7 - Nanoparticle-based vaccines: opportunities and limitations. In:Nanopharmaceuticals Shegokar R, , editor. Elsevier. (2020). p. 135–50. doi: 10.1016/B978-0-12-817778-5.00007-5

Crossref Full Text | Google Scholar

275. Qie Y, Yuan H, von Roemeling CA, Chen Y, Liu X, Shih KD, et al. Surface modification of nanoparticles enables selective evasion of phagocytic clearance by distinct macrophage phenotypes. Sci Rep. (2016) 6:26269. doi: 10.1038/srep26269

PubMed Abstract | Crossref Full Text | Google Scholar

276. Li Y, Tenchov R, Smoot J, Liu C, Watkins S, Zhou Q. A comprehensive review of the global efforts on COVID-19 vaccine development. ACS Central Sci. (2021) 7:512–33. doi: 10.1021/acscentsci.1c00120

PubMed Abstract | Crossref Full Text | Google Scholar

277. Mohsen MO, Zha L, Cabral-Miranda G, Bachmann MF. Major findings and recent advances in virus–like particle (VLP)-based vaccines. Semin Immunol. (2017) 34:123–32. doi: 10.1016/j.smim.2017.08.014

PubMed Abstract | Crossref Full Text | Google Scholar

278. Lu Y, Clark-Deener S, Gillam F, Heffron CL, Tian D, Sooryanarain H, et al. Virus-like particle vaccine with B-cell epitope from porcine epidemic diarrhea virus (PEDV) incorporated into hepatitis B virus core capsid provides clinical alleviation against PEDV in neonatal piglets through lactogenic immunity. Vaccine. (2020) 38:5212–8. doi: 10.1016/j.vaccine.2020.06.009

PubMed Abstract | Crossref Full Text | Google Scholar

279. Ward BJ, Gobeil P, Séguin A, Atkins J, Boulay I, Charbonneau PY, et al. Phase 1 randomized trial of a plant-derived virus-like particle vaccine for COVID-19. Nat Med. (2021) 27:1071–8. doi: 10.1038/s41591-021-01370-1

PubMed Abstract | Crossref Full Text | Google Scholar

280. Yilmaz IC, Ipekoglu EM, Bulbul A, Turay N, Yildirim M, Evcili I, et al. Development and preclinical evaluation of virus-like particle vaccine against COVID-19 infection. Allergy. (2022) 77:258–70. doi: 10.1111/all.15091

PubMed Abstract | Crossref Full Text | Google Scholar

281. Dhakal S, Hiremath J, Bondra K, Lakshmanappa YS, Shyu DL, Ouyang K, et al. Biodegradable nanoparticle delivery of inactivated swine influenza virus vaccine provides heterologous cell-mediated immune response in pigs. J Controll Release. (2017) 247:194–205. doi: 10.1016/j.jconrel.2016.12.039

PubMed Abstract | Crossref Full Text | Google Scholar

282. Karch CP, Burkhard P. Vaccine technologies: from whole organisms to rationally designed protein assemblies. Biochem Pharmacol. (2016) 120:1–14. doi: 10.1016/j.bcp.2016.05.001

PubMed Abstract | Crossref Full Text | Google Scholar

283. Cavalcanti IDL, Cajubá de. Britto Lira Nogueira M. Pharmaceutical nanotechnology: which products are been designed against COVID-19? J Nanoparticle Res. (2020) 22:276. doi: 10.1007/s11051-020-05010-6

PubMed Abstract | Crossref Full Text | Google Scholar

284. Thalhauser S, Peterhoff D, Wagner R, Breunig M. Presentation of HIV-1 envelope trimers on the surface of silica nanoparticles. J Pharm Sci. (2020) 109:911–21. doi: 10.1016/j.xphs.2019.10.059

PubMed Abstract | Crossref Full Text | Google Scholar

285. Mateu Ferrando R, Lay L, Polito L. Gold nanoparticle-based platforms for vaccine development. Drug Discov Today: Technol. (2020) 38:57–67. doi: 10.1016/j.ddtec.2021.02.001

PubMed Abstract | Crossref Full Text | Google Scholar

286. Sekimukai H, Iwata-Yoshikawa N, Fukushi S, Tani H, Kataoka M, Suzuki T, et al. Gold nanoparticle-adjuvanted S protein induces a strong antigen-specific IgG response against severe acute respiratory syndrome-related coronavirus infection, but fails to induce protective antibodies and limit eosinophilic infiltration in lungs. Microbiol Immunol. (2020) 64:33–51. doi: 10.1111/1348-0421.12754

PubMed Abstract | Crossref Full Text | Google Scholar

287. Qiao L, Chen M, Li S, Hu J, Gong C, Zhang Z, et al. A peptide-based subunit candidate vaccine against SARS-CoV-2 delivered by biodegradable mesoporous silica nanoparticles induced high humoral and cellular immunity in mice. Biomater Sci. (2021) 9:7287–96. doi: 10.1039/D1BM01060C

PubMed Abstract | Crossref Full Text | Google Scholar

288. Khalaj-Hedayati A, Chua CLL, Smooker P, Lee KW. Chapter 2 - Universal influenza vaccine technologies and recombinant virosome production. In:Gurtler V, , editor. Methods in Microbiology. Amsterdam: Academic Press (2022). p. 45–89. doi: 10.1016/bs.mim.2022.04.001

Crossref Full Text | Google Scholar

289. Ali H, Akbar M, Iqbal B, Ali F, Sharma NK, Kumar N, et al. Virosome: an engineered virus for vaccine delivery Saudi. Pharma J. (2023) 31:752–64. doi: 10.1016/j.jsps.2023.03.016

PubMed Abstract | Crossref Full Text | Google Scholar

290. Fernandes B, Castro R, Bhoelan F, Bemelman D, Gomes RA, Costa J, et al. Insect cells for high-yield production of SARS-CoV-2 spike protein: building a virosome-based COVID-19 vaccine candidate. Pharmaceutics. (2022) 14:854. doi: 10.3390/pharmaceutics14040854

PubMed Abstract | Crossref Full Text | Google Scholar

291. Abhyankar MM, Mann BJ, Sturek JM, Brovero S, Moreau GB, Sengar A, et al. Development of COVID-19 vaccine using a dual Toll-like receptor ligand liposome adjuvant. NPJ Vacc. (2021) 6:137. doi: 10.1038/s41541-021-00399-0

PubMed Abstract | Crossref Full Text | Google Scholar

292. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines — a new era in vaccinology. Nat Rev Drug Discov. (2018) 17:261–79. doi: 10.1038/nrd.2017.243

PubMed Abstract | Crossref Full Text | Google Scholar

293. Wilson B, Geetha KM. Lipid nanoparticles in the development of mRNA vaccines for COVID-19. J Drug Deliv Sci Technol. (2022) 74:103553. doi: 10.1016/j.jddst.2022.103553

PubMed Abstract | Crossref Full Text | Google Scholar

294. Hussain A, Yang H, Zhang M, Liu Q, Alotaibi G, Irfan M, et al. mRNA vaccines for COVID-19 and diverse diseases. J Controll Release. (2022) 345:314–33. doi: 10.1016/j.jconrel.2022.03.032

PubMed Abstract | Crossref Full Text | Google Scholar

295. Li C, Yang T, Weng Y, Zhang M, Zhao D, Guo S, et al. Ionizable lipid-assisted efficient hepatic delivery of gene editing elements for oncotherapy. Bioact Mater. (2022) 9:590–601. doi: 10.1016/j.bioactmat.2021.05.051

PubMed Abstract | Crossref Full Text | Google Scholar

296. Ho W, Gao M, Li F, Li Z, Zhang XQ, Xu X. Next-generation vaccines: nanoparticle-mediated DNA and mRNA delivery. Adv Healthc Mater. (2021) 10:2001812. doi: 10.1002/adhm.202001812

PubMed Abstract | Crossref Full Text | Google Scholar

297. Samaridou E, Heyes J, Lutwyche P. Lipid nanoparticles for nucleic acid delivery: current perspectives. Adv Drug Deliv Rev. (2020) 154–5:37–63. doi: 10.1016/j.addr.2020.06.002

PubMed Abstract | Crossref Full Text | Google Scholar

298. Sebastiani F, Yanez Arteta M, Lerche M, Porcar L, Lang C, Bragg RA, et al. Apolipoprotein E binding drives structural and compositional rearrangement of mRNA-containing lipid nanoparticles. ACS Nano. (2021) 15:6709–22. doi: 10.1021/acsnano.0c10064

PubMed Abstract | Crossref Full Text | Google Scholar

299. DeFrancesco L. Whither COVID-19 vaccines? Nat Biotechnol. (2020) 38:1132–45. doi: 10.1038/s41587-020-0697-7

PubMed Abstract | Crossref Full Text | Google Scholar

300. Cascella M, Rajnik M, Aleem A, Dulebohn SC, Di Napoli R. Features, evaluation, and treatment of coronavirus (COVID-19) (2023). StatPearls.

Google Scholar

301. WHO COVID-19 Dashboard - Variants. Geneva: WHO (2025).

Google Scholar

302. Satapathy P. Global spread of COVID-19's JN.1 variant: implications and public health responses. N Microbes N Infect. (2024) 57:101225. doi: 10.1016/j.nmni.2024.101225

PubMed Abstract | Crossref Full Text | Google Scholar

303. Benucci M, Bernardini P, Coccia C, De Luca R, Levani J, Economou A, et al. JAK inhibitors and autoimmune rheumatic diseases. Autoimmun Rev. (2023) 22:103276. doi: 10.1016/j.autrev.2023.103276

PubMed Abstract | Crossref Full Text | Google Scholar

304. Manna S, Samal P, Basak R, Mitra A, Roy AK, Kundu R, et al. Amentoflavone and methyl hesperidin, novel lead molecules targeting epitranscriptomic modulator in acute myeloid leukemia: in silico drug screening and molecular dynamics simulation approach. J Mol Model. (2023) 29:9. doi: 10.1007/s00894-022-05407-1

PubMed Abstract | Crossref Full Text | Google Scholar

305. Lingwan M, Shagun S, Pahwa F, Kumar A, Verma DK, Pant Y, et al. Phytochemical rich Himalayan Rhododendron arboreum petals inhibit SARS-CoV-2 infection in vitro. J Biomol Struct Dyn. (2023) 41:1403–13. doi: 10.1080/07391102.2021.2021287

PubMed Abstract | Crossref Full Text | Google Scholar

306. Park JW, Lagniton PNP, Liu Y, Xu R-H. mRNA vaccines for COVID-19: what, why and how. Int J Biol Sci. (2021) 17:1446–60. doi: 10.7150/ijbs.59233

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: COVID-19, etiology, pathogenicity, pathophysiology, synthetic, natural products

Citation: El Zawily A, Eckert S, Adajar R, Wagih N, Elmaidomy AH, Helmy AM, Mustafa M, Elshorbagi M, Ghali E, Fadl RG, Bodem J, Abdelmohsen UR and Zaki MYW (2025) Comprehensive review on COVID-19: etiology, pathogenicity, and treatment. Front. Med. 12:1569013. doi: 10.3389/fmed.2025.1569013

Received: 03 February 2025; Accepted: 25 August 2025;
Published: 23 September 2025.

Edited by:

Sunil Dhiman, Defence Research & Development Establishment (DRDE), India

Reviewed by:

Shetty Ravi Dyavar, Adicet Bio, Inc., United States
Agnieszka Kwiatek, University of Warsaw, Poland

Copyright © 2025 El Zawily, Eckert, Adajar, Wagih, Elmaidomy, Helmy, Mustafa, Elshorbagi, Ghali, Fadl, Bodem, Abdelmohsen and Zaki. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Usama Ramadan Abdelmohsen, dXNhbWEucmFtYWRhbkBtdS5lZHUuZWc=; Amr El Zawily, YWVsemF3aWxAdW9ndWVscGguY2E=

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.