Unmasking vulnerabilities in the age of COVID‑19 (Review)
- Authors:
- Published online on: October 31, 2024 https://doi.org/10.3892/wasj.2024.290
- Article Number: 2
-
Copyright : © Mustafa et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].
Abstract
1. Introduction
Severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2), which belongs to the coronaviridae family is the source of the current coronavirus disease 2019 (COVID-19) pandemic, which continues to affect humanity. The real-time observation of ongoing evolutionary processes has provided a marked understanding of SARS-CoV-2 diversification. Numerous variations have emerged as a result of this diversification, each set apart by unique traits, such as immunological evasion, severity and transmissibility. Changes in immune profiles, human migration and infected individuals are all part of the complex evolutionary path that is intimately connected to ecological dynamics and the events of transmission (1). Among the 180 identified species of ribonucleic acid (RNA) viruses capable of infecting humans, an average of two new species emerge each year. RNA viruses have extensively spread among humans, other mammals and occasionally birds, across both epidemiological and evolutionary timelines. Notably, 89% of human-infective species are zoonotic, and a considerable proportion of the remaining species trace their origins back to zoonotic sources. The pace at which mutations are created and propagated across populations is the most critical factor in viral evolution (2). Natural selection also helps to fix favorable mutations and improve transmissibility (3). However, viral evolution becomes complex when viruses reproduce and develop inside humans, while adapting to effective human-to-human transmission. As viral lineages evolve, antigenically distinct strains may emerge at higher organizational levels (4). The present review aimed to provide insight into the evolutionary dynamics of SARS-CoV-2 across various scales. This encompasses examining the stages of the COVID-19 pandemic, identifying crucial factors influencing the evolution of the virus, exploring hypotheses surrounding the emergence of statistically significant variants, and contemplating potential evolutionary pathways that could affect public health in the future. Considering the substantial role of SARS-CoV-2 in triggering the COVID-19 pandemic, a comprehensive investigation into the infection and its repercussions for public health is essential. The present review also delves into the transmission of SARS-CoV-2 from patient to host, the utilization of mathematical models for predicting the risk of viral aerosol/droplet transmission, potential pathways for viral entry into the human host and the cellular mechanisms underlying these processes.
In addition, the present review highlights the COVID-19 clinical symptoms and available diagnostic approaches for detecting the virus. The requirement for effective treatment techniques, such as vaccine development and medication repurposing, is emphasized herein. Given the considerable amount of studies on COVID-19 and the available literature, it appears difficult to address each element. The present review aimed to provide a comprehensive discussion of diverse facets concerning the COVID-19 outbreak. It covers a wide range of topics, such as preventative measures against the virus, clinical characteristics of symptomatic and asymptomatic individuals, estimations of the infection and incubation periods, the immune responses that the virus elicits in humans, and the association between pre-existing comorbidities and COVID-19-associated mortality. Furthermore, the present review provides a historical framework for understanding pandemics, tracing their evolution from confined outbreaks to global epidemics, starting in the 16th and 19th centuries. It delves into zoonotic origins, elucidating the transmission of zoonoses from animals to humans, with illustrative examples, such as human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS), Ebola and historical influenza strains. SARS-CoV-2 exhibits significant genetic similarities to pangolin coronaviruses and bat betacoronaviruses, indicating that the ongoing COVID-19 pandemic has its origins in an animal reservoir (5). As the battle against COVID-19 continues, the acquisition of knowledge and understanding remains indispensable in formulating efficacious strategies to safeguard global public health.
2. Emergence and spread of COVID-19
Several pneumonia cases with an unclear cause emerged in late December, 2019, in Wuhan, Hubei Province, China (6). The afflicted individuals exhibited clinical signs of fever, cough, dyspnea, chest pain and bilateral lung infiltration, symptoms of viral pneumonia, which were comparable to those in SARS and Middle East respiratory syndrome (MERS) (7). The Huanan Seafood Wholesale Market, a wet market in downtown Wuhan known for selling seafood and live animals, including poultry and wildlife, was linked to the majority of the initial cases (8). On December 8, 2019, the earliest case was recorded (9).
The World Health Organization (WHO) was formally notified that the Wuhan Municipal Health Commission reported an unknown pneumonia outbreak on December 31. Independent Chinese scientific teams revealed a novel betacoronavirus as the cause of this newly discovered disease (10). The first genome sequence of the novel coronavirus was made available on January 10. The outbreak coincided with the Lunar New Year celebrations, which led to more individuals traveling and spreading the virus to additional Hubei Province cities, and ultimately to other regions of China (11). The escalation in severity led the WHO to declare the COVID-19 outbreak as a public health emergency of international concern on January 30, 2020(12). The WHO officially designated the illness as COVID-19 on February, 11 2020(13). A schematic diagram of the timeline of these events is presented in Fig. 1. China imposed strict public health measures, such as a city-wide lockdown of Wuhan on January 23, 2020 with travel and transportation restrictions, to contain the outbreak (14). The high transmissibility of the virus and global travel contributed to large clusters of infections being reported in numerous countries. Consequently, on March 11, 2020, the WHO formally declared the COVID-19 outbreak to be a pandemic (15,16). China was able to contain the virus relatively well; however, the number of cases in the USA and Europe increased rapidly (17).
As of August 20, 2024, the USA is experiencing a high prevalence of the SARS-CoV-2 Omicron variants KP.2, KP.2.3, KP.3, KP.3.1.1 and LB.1. The Center for Disease Control and Prevention indicate that KP.3.1.1 is expected to comprise ~37% of new COVID-19 cases in the USA. The estimated percentage of illnesses caused by KP.2.3 remains at 14.4%, similar to the previous 2-week period, and the estimated rate of diseases caused by LB.1 also remains at 14.1%. JN.1 has been reported by 115 countries, rendering it the most reported VOI, representing 90.3% of sequences in week 9, up from 89.4% in week 6. The parent lineage of JN.1, BA.2.86, decreased and was responsible for 2.2% of sequences in week 9, down from 3.0% in week 6 (https://covid.cdc.gov/covid-data-tracker/#variant-summary; https://www.who.int/publications/m/item/covid-19-epidemiological-update-15-march-2024).
3. SARS and COVID-19: Similarities and differences
There are notable similarities between the clinical manifestations and modes of transmission of the 2019 COVID-19 and SARS virus. Both infections have the potential to manifest as rapidly progressing pneumonia. It appears that the primary mode of transmission for both is infectious respiratory droplets that are released from mucosal membranes (Table I). The viruses exhibit comparable stability and degradation in aerosols and on various surfaces (18). According to researchers, both viruses can live for up to 2 days on stainless steel and 3 days on plastic, and their viral titers on both surfaces exhibit comparable decay patterns (19-21). Both SARS and COVID-19 appear to have a median incubation period of 4 to 7 days from first exposure to the start of symptoms. Furthermore, according to research, the maximum incubation time for both might be up to 14 days (22-24). This longer incubation time adds to the difficulty of preventing the spread of these illnesses. Despite these similarities, it is important to emphasize that SARS and COVID-19 are caused by different viruses and are members of separate coronavirus subfamilies. In summary, whereas SARS and COVID-19 share clinical signs and transmission characteristics, they are caused by separate viruses and have distinct characteristics that distinguish them as distinct causative agents. Understanding these similarities and differences appears critical for successful epidemic management and prevention measures. The incubation time and length of viral shedding are critical for determining the risk of transmission, adopting isolation and quarantine measures, and developing effective antiviral therapies for patients. According to recent epidemiological research, the typical period of COVID-19 virus shedding is ~20 days, with some survivors shedding for as long as 37 days (25). By contrast, viral RNA remains detectable in non-survivors until mortality. Patients with severe COVID-19 infection may suffer viral shedding for a median of 31 days after the disease begins (26).
Betacoronaviruses and alphacoronaviruses have natural hosts in bats. RaTG13, a bat coronavirus isolated from Rhinolophus affinis in Yunnan province, China, is the closest known match to SARS-CoV-2 to date (27). RaTG13 and SARS-CoV-2 share 96.2% of the full-length genome sequence, demonstrating a strong genetic similarity (28,29). The fact that SARS-CoV-2 and RaTG13 share >90% of their genome's sequence, including the variable spike glycoprotein (S glycoprotein) and open reading frame (ORF)8 regions, is particularly notable (28). Their close association is highlighted by phylogenetic analysis, which lends credence to the theory that bats are the original host of SARS-CoV-2. SARS-CoV-2 and ‘RmYN02’, a recently discovered coronavirus found in a Yunnan Rhinolophus malayanus bat, share 93.3% of their genome (29). Notably, it shares a longer 1ab gene with SARS-CoV-2 with 97.2% identity, higher than RaTG13(30). Furthermore, ZC45 and ZXC21, two additional bat coronaviruses that were previously discovered in eastern Chinese Rhinolophus pusillus bats, are members of the SARS-CoV-2 lineage within the Sarbecovirus subgenus (31). These findings highlight the wide range of bat coronaviruses that are strongly associated with SARS-CoV-2, indicating that bats may be the virus's possible hosts. Recent investigations have revealed that the genetic diversity observed in SARS-CoV-2 and its related bat coronaviruses stems from over 20 years of sequence evolution (31,32). Consequently, it is incorrect to categorize these bat coronaviruses as the immediate progenitors of SARS-CoV-2, despite being likely evolutionary ancestors.
Pangolins are another possible animal host connected to SARS-CoV-2. Between 2017 and 2019, several viruses related to SARS-CoV-2 were discovered in the tissues of pangolins (33). These pangolin viruses are from two distinct sub-lineages and were independently traced in the provinces, of Guangxi and Guangdong (34-37). Pangolins linked to various smuggling incidents have been found to have SARS-CoV-2-related coronavirus infections, suggesting that these animals may serve as hosts for the viruses (38). Pangolins infected with coronaviruses display clinical symptoms and histological changes, such as multiple organ infiltration of inflammatory cells and interstitial pneumonia, in contrast to bats, which typically carry the virus without obvious damage (39).
Emerging coronaviruses that are derived from bats require an intermediate host to proliferate. For example, dromedary camels and palm civets served as intermediary hosts for SARS-CoV and MERS-coronavirus (MERS-CoV), respectively (40). The viruses harbored by these hosts share a genome sequence identity of >99% with the corresponding viruses in humans (41). The role of an intermediary host in the transmission of the SARS-CoV-2 virus, which is accountable for the COVID-19 pandemic, is under scrutiny and remains unclear. Pangolin coronaviruses exhibit only a 92% genomic identity with SARS-CoV-2, despite displaying a marked similar receptor-binding domain (RBD) (42). Consequently, it is challenging to definitively ascertain whether pangolins acted as the intermediate host for SARS-CoV-2 or if they were directly implicated in the emergence of the virus. The animal source of SARS-CoV-2 is presently poorly understood, with limited knowledge available on this aspect. The reservoir hosts of the virus have yet not been identified, nor it has been determined if an intermediate host was involved in the transmission of the virus to humans. Significantly, the discovery of pangolin coronaviruses, RaTG13, and RmYN02 implies that SARS-CoV-2-like coronaviruses are prevalent in animals (43-45).
In addition to wildlife, research has explored the susceptibility of domesticated and laboratory animals to SARS-CoV-2 infection. Experimental findings have demonstrated that SARS-CoV-2 can effectively replicate in cats and ferrets, particularly in the upper respiratory tract (46). Conversely, dogs, pigs, chickens and ducks have exhibited immunity to the virus (47). Notably, minks have been observed to contract SARS-CoV-2, as evidenced by an outbreak on mink farms in The Netherlands, leading to severe cases of respiratory distress and interstitial pneumonia (48). Although devoid of symptoms, two dogs in Hong Kong tested positive for spontaneous SARS-CoV-2 infection through serological and virological tests (49). Similarly, the analysis of blood samples from cats in Wuhan revealed the presence of neutralizing antibodies against SARS-CoV-2, confirming the infection in cat populations. However, the possibility of transmission from cats to humans remains uncertain (50). Ongoing comprehensive research and surveillance on animal susceptibility aim to provide a deeper understanding of potential hosts and transmission dynamics of the virus.
4. Comparative insight into SARS-CoV-2: Infectiousness, transmission and evolution
The virus accountable for acute respiratory illness, SARS-CoV-2, belongs to the coronavirus family and carries a non-segmented genome composed of positive-sense, single-stranded RNA enveloped by the viral capsid (51). Coronaviruses (CoVs) are categorized into four genera: α, β, γ and δ-CoV (52). While α- and β-CoV predominantly infect mammals, they can also affect birds. Human-infecting coronaviruses include HCoV-229E, SARS-CoV, HCoV-OC43, HCoV-NL63, MERS-CoV and HCoV-HKU1(53). Infections caused by HCoV-229E, HCoV-NL63, HCoV-HKU1 and HCoV-OC43 typically result in mild respiratory symptoms. By contrast, SARS-CoV and MERS-CoV can lead to severe respiratory disease, occasionally resulting in death due to multiple organ failure (54). SARS-CoV-2 shares notable similarities (>85%) with bat-derived SARS-like coronaviruses identified as bat-SL-CoVZC45 and bat-SL-CoVZXC21(55). Compared to SARS-CoV and MERS-CoV, it demonstrates ~79 and 50% homology, respectively (56). This evidence and phylogenetic research strongly indicate that SARS-CoV-2 originated in bats and potentially transmitted to humans through an unidentified intermediate host species. The genomic structure, encoded structural and non-structural proteins, and the primary host of SARS-CoV-2 are illustrated in Fig. 2.
The pathogenesis of SARS-CoV-2 involves a complex interplay of viral and host factors. As an enveloped positive-sense single-stranded RNA virus, the genomic structure of SARS-CoV-2 comprises a significant portion (two thirds) dedicated to an ORF (ORF 1a/b), encoding 16 non-structural proteins crucial for replication. The remaining section of the genome encodes essential structural proteins (S glycoprotein, small envelope protein, matrix protein and nucleocapsid protein) and accessory proteins with functions still under investigation. The S glycoprotein, essential for host cell entry, binds to the angiotensin-converting enzyme 2 (ACE2) receptor. However, the precise mechanism of membrane invagination for SARS-CoV-2 endocytosis remains unclear. Host factors, particularly ACE2 expression, influence viral tropism. The elderly and individuals with underlying health conditions are more susceptible to severe infections, partly due to age-related immune system changes and comorbidities. Host immune responses, both innate and adaptive, play a crucial role, and dysregulated responses can contribute to disease severity. Additionally, genetic factors contribute to interindividual variability in susceptibility and disease outcomes. A comprehensive understanding of these viral and host elements is crucial for developing effective therapeutic interventions and vaccines against SARS-CoV-2. Ongoing research continues to unveil additional details about the intricate virus-host interactions shaping the pathogenesis of COVID-19. Without a doubt, expressive experimentation has shown that the virus infects people by attaching itself to respiratory system-expressed ACE2 receptors (57,58). Overall, the findings from several investigations demonstrate that SARS-CoV-2 is extremely infectious, with viral shedding commencing before symptoms develop and the virus spreading through many channels (15-17,59). Controlling the spread of the disease is a main concern for public health initiatives.
Although SARS-CoV-2 is less severe in terms of morbidity and mortality than MERS and SARS, it is more contagious. COVID-19 has a much lower mortality rate of 3.4% compared to 9.6 and 35% for SARS and MERS, respectively. COVID-19 primarily spreads through person-to-person contact, particularly between close friends and family members (59). Numerous studies have demonstrated the critical role symptomatic individuals play in COVID-19 transmission, mainly through respiratory droplet expulsion from actions, such as coughing or sneezing. On the other hand, nosocomial transmission was primarily responsible for the spread of MERS-CoV and SARS-CoV among healthcare personnel (60). In MERS-CoV outbreaks, medical staff was responsible for 62-79% of cases, whereas in the SARS case, they accounted for 33-42% of cases. The most likely ways for a virus to spread are through direct contact with the host or interactions with an unidentified intermediate carrier (61).
The SARS-CoV-2 virus changes in a variety of ways as it grows and spreads among the population. In December, 2020, a noteworthy variant, VUI-202012/01, was examined due to 17 distinct alterations or mutations in its DNA. Since the discovery of SARS-CoV-2 in 2019, thousands of mutations have already manifested in its genome (62). As the pandemic continues, the continual mutation process in the population may result in the production of immunologically relevant mutations, thereby affecting vaccination effectiveness. These mutations are resulting in novel viral combinations. The COVID-19 genomics UK consortium (COG-UK) has conducted extensive epidemiological and virological investigations in response to the significant surge in COVID-19 cases recently observed in the UK, particularly in South East England (63). A novel variant was identified in viral genome sequences, forming a distinct phylogenetic grouping. This variant is distinguished by multiple spike protein mutations (deletion 69-70, deletion 144, N501Y, A570D, D614G, P681H, T716I, S982A and D1118H), accompanied by alterations in other genomic regions (64). Although viral mutations are normal, preliminary studies have indicated that this variant in the UK may be critical for increased transmissibility and is projected to possibly raise the reproductive number by 0.4 or more (65). Notably, this new variety evolved during a period of increased family and social gatherings. However, there is no indication that it causes more severe infections than other variations.
5. Role of ACE2 receptor, an angiotensin-converting enzyme
SARS-CoV-2 gains entry into the human host through receptor-mediated endocytosis, a mechanism through which viruses bind to specific receptors on the cell surface of the host, facilitating entry. The RBD of the virus establishes a connection with the appropriate receptor on the host cell, enabling entry. Both SARS-CoV and SARS-CoV-2 utilize the ACE2 receptor to infect cells. Previous research has shown that the S protein of SARS-CoV exhibits a strong affinity for the ACE2 receptor, serving as the entry point for the virus into host cells (66). A schematic diagram depicting the fusion of the virus with the host receptor is presented in Fig. 3.
The entry of SARS-CoV-2 into host cells is also mediated by S protein priming by transmembrane protease serine-2 (TMPRSS2). This priming event is crucial for the fusion of the viral envelope with the host cell membrane, enabling subsequent viral entry. Therefore, the coordinated interplay between the ACE2 receptor and TMPRSS2 is essential for the efficient entry of SARS-CoV-2 into the host environment. It is noteworthy that TMPRSS2 exhibits a higher expression and broader distribution compared to ACE2 receptors, suggesting that ACE2 may function as a limiting factor during the initial infection phase. While TMPRSS2 is a key component for viral entry, alternate proteases, such as cathepsin B/L, may act as substitutes for TMPRSS2. Hence, the simultaneous inhibition of these proteases becomes crucial in preventing cellular entry.
The structural characteristics of the S proteins of SARS-CoV and SARS-CoV-2 facilitate the entry of the latter into cells (67). Studies involving human HeLa cells and animals with and without ACE2 expression support the involvement of ACE2 receptors in the cellular entry of the SARS-CoV-2 virus, particularly the Wuhan strain (68,69). Research on the SARS-CoV-2 infection of BHK21 cells has indicated higher infection rates when transfected with human and bat ACE2 receptors compared to BHK21 cells lacking ACE2 expression (70,71). Biophysical and structural data suggest that the ACE2 binding affinity of the SARS-CoV-2 S protein ectodomain is significantly greater than that of the SARS-CoV S protein by a ratio of 10:20(72). This difference is considered to contribute to the variance in contagiousness between SARS-CoV-2 and SARS-CoV. Although the ACE2 and ACE-1 receptors share similarities, the ACE2 receptor has a smaller active site and a smaller binding pocket with different amino acids, making it resistant to typical ACE inhibitors, such as lisinopril, enalapril, and ramipril (73).
Furthermore, there is no evidence to suggest that angiotensin receptor blockers (ARBs), such as losartan, disrupt the activity of ACE2. TMPRSS2, identified as a type II transmembrane protease, consists of distinct domains, including an intracellular N-terminal domain, a transmembrane domain, an extracellularly extending stem region, and a C-terminal domain facilitating its serine protease (SP) function (74). The serine protease activity relies on a catalytic triad, comprised of His296, Asp345 and Ser441, responsible for cleaving basic amino acid residues, particularly lysine or arginine residues, aligning with its role in cleaving the S1/S2 site in SARS-CoV-2(75).
While TMPRSS2 has been recognized for its involvement in prostate cancer and viral infections, such as influenza, SARS and MERS (76), it has recently gained attention from drug developers. Multiple studies are underway to uncover strategies aimed at reducing TMPRSS2 expression or blocking its activity in host cell membranes, with the ultimate goal of inhibiting SARS-CoV-2 entry into host cells (77,78).
6. Diagnostic and therapeutic approaches, and strategies to inhibit viral entry
The molecular detection of SARS-CoV-2 nucleic acid is the most accurate diagnostic approach (79). Various commercially available kits for viral nucleic acid detection target different genes, including ORF1ab (containing RdRp), N, E or S (80). The detection time may vary from a few minutes to several hours depending on the technology utilized. Although SARS-CoV-2 has been detected in throat swabs, posterior oropharyngeal saliva, nasopharyngeal swabs, sputum and bronchial fluid, the viral load is notably higher in samples from the lower respiratory tract (81). Viral nucleic acid has also been detected in intestinal and blood samples, even in cases where respiratory tests yielded negative results. The viral load may decrease from its peak at the onset of the illness, potentially leading to false negatives when using oral swabs (81). It is advisable to employ multiple detection techniques to confirm a COVID-19 diagnosis.
To address the issue of false negatives, alternative detection approaches have been utilized. Therefore, for individuals with a robust clinical suspicion of COVID-19 despite an initial negative nucleic acid screening, a combination of CT scans and repeated swab testing has been recommended. Serological assays that identify antibodies to the N or S protein of SARS-CoV-2 could complement molecular diagnosis, particularly in the latter stages of the illness or for retrospective research (76,82,83). The magnitude and duration of immunological responses are still unknown, and the sensitivity and specificity of existing serological assays vary. When selecting and interpreting serological testing, all these factors should be taken into consideration, possibly even extending to future assays for T-cell responses (84).
Currently, neither COVID-19 nor specific antivirals that target SARS-CoV-2 have the potential to combat the disease. However, several treatments have shown some promise. Manufacturers and researchers are undertaking large clinical studies to examine new COVID-19 therapy options.
The pharmaceutical interventions available for COVID-19 therapy can be divided into several groups: Immunotherapy, cellular therapy, antiviral and other drugs. Immunotherapy mainly includes; immunoglobulins, interferons, convalescent plasma, and monoclonal therapy. Human immunoglobulin is tested in patients with pneumonia with COVID-19 and there are 478 clinical trials concerning immunoglobulins; 18 of these trials have been completed (https://www.clinicaltrials.gov/search?term=immunoglobulin&cond=Covid19&viewType=Table).
There are 87 clinical trials underway about interferons against COVID-19; of these, six clinical trials have been completed (https://www.clinicaltrials.gov/search?term=interferon&cond=Covid19&viewType=Table). In addition, 140 clinical trials examining the efficacy and safety of convalescent plasma for COVID-19 and, of these, nine trials have been completed (https://www.clinicaltrials.gov/search?term=convalescent%20plasma&cond=Covid19&viewType=Table).
Currently, 47 clinical trials are being conducted for monoclonal antibodies; none of have been completed (https://www.clinicaltrials.gov/search?term=monoclonal%20antibody&cond=Covid19&viewType=Table).
Cellular therapy primarily focuses on mesenchymal stem cells (MSCs) and natural killer (NK) cells. MSCs are potent anti-inflammatory and immunomodulatory tools of the immune system. MSCs can reduce the burden of lung injury and respiratory distress by preventing leakage of immune cells and proinflammatory cytokines to the pulmonary tissue. Of note, there are 55 ongoing clinical trials and two trials have been completed (https://www.clinicaltrials.gov/search?term=mesenchymal%20stem%20cell&cond=Covid19&viewType=Table).
As regards NK cells, they can identify the infected cells, mediate antibody-dependent cytotoxicity and maintain immunological homeostasis. A total of 28 clinical trials are underway on NK cells (https://www.clinicaltrials.gov/search?term=natural%20killer%20cell&cond=Covid19&viewType=Table). There are also ~84 clinical trials conducted with other drugs, and of these, 9 trials have been completed (https://clinicaltrials.gov/search?term=chloroquine&cond=Covid19&viewType=Table).
Potential antiviral targets for the treatment of COVID-19 are depicted in Fig. 3. A crucial strategy in combatting SARS-CoV-2 infection is to hinder viral entry. ACE2 exists in membrane-bound ACE2 (mACE2), located in the gallbladder, heart, intestines, kidneys and testes (85). The virus uses human proteases as entry activators to break through host cells through membrane fusion, and it uses ACE2 as a receptor. Treatments aimed at this entry mechanism have the potential for treating COVID-19. Umifenovir, also known as Arbidol, is a drug approved for treating respiratory viral infections and influenza in China and Russia. Its mechanism of action involves preventing membrane fusion by interfering with the interaction between the S protein and ACE2(86). In vitro studies have demonstrated its efficacy against SARS-CoV-2; clinical data suggest that it may present a more effective treatment for COVID-19 when compared to lopinavir and ritonavir (87-91).
One notable drug that shows promise is camostat mesylate, which is licensed in Japan for the treatment of post-operative reflux esophagitis and pancreatitis (92). Previous studies have demonstrated the ability of camostat mesylate to inhibit TMPRSS2 activity and protect mice from fatal SARS-CoV infection (93). Recent studies have further indicated that camostat mesylate can inhibit the entry of SARS-CoV-2 into human lung cells (88). This suggests potential utility as an antiviral drug against SARS-CoV-2 in the future, although further clinical data is required to confirm its effectiveness (92-95).
Other drugs used to treat autoimmune diseases and prevent malaria, such as chloroquine and hydroxychloroquine, may also influence SARS-CoV-2 entry. They function by preventing membrane fusion by raising endosomal pH, interfering with the interaction between virus and host receptor, and inhibiting the glycosylation of cellular receptors (87). As regards their effectiveness in treating COVID-19, there remains a lack of scientific consensus. Despite concerns about an increased risk of cardiac arrest in treated patients, two clinical investigations found no correlation between these medications and patient mortality rates (96,97). On June 15, 2020, due to documented adverse events, the US Food and Drug Administration (FDA) revoked the emergency use authorization for chloroquine and hydroxychloroquine in COVID-19 therapy (98).
Another therapeutic approach involves the use of soluble recombinant human ACE2 (hACE2), specific monoclonal antibodies, or fusion inhibitors targeting the SARS-CoV-2 S protein to prevent its binding to the ACE2 receptor (99). Examples of replication inhibitors include remdesivir (GS-5734), favilavir (T-705), ribavirin, lopinavir and ritonavir. The remaining three agents act on RdRp, except for lopinavir and ritonavir, which inhibit 3CLpro. Potential antiviral targets for COVID-19 treatment are illustrated in Fig. 4. However, further clinical research is required to evaluate the effectiveness and safety of these approaches.
Novel diagnostic approaches are currently aimed at detecting COVID-19. Developing accessible point-of-care diagnostic devices customized for low-resource settings remains an urgent need. This can be accomplished through workflows that do not require cell lysis, bioassays that are increasingly sensitive and robust, improved methods for sample collection and processing that minimize risks to healthcare workers, and kits that are ready-to-use and do not require technical expertise, among other potential innovations. RT-LAMP and CRISPR/Cas-based methods, such as Specific High-Sensitivity Enzymatic Reporter UnLOCKing (SHERLOCK) and DNA Endonuclease-Targeted CRISPR Trans Reporter (DETECTR) are significant advancements that could be considered as promising alternatives (100,101). As regards portable diagnostic devices, few technologies hold as much promise as paper-based microfluidics (102). Natural remedies, such as Withania somnifera and Tinospora cordifolia have been tested on experiment models of COVID-19; research has shown their potency to limit the deterioration of the health of patients by reducing inflammation (99). Furthermore, the researchers are investigating the repurposing of drugs, such as procaine, which is an antiviral agent. It has demonstrated the in vitro inhibition of SARS-CoV-2, facilitating novel approaches for the treatment of the disease (103).
The implementation of these strategies requires vast capital investment. In light of concerns about a potential economic downturn and financial breakdown, resilient and effective leadership needs to emerge in healthcare, business, government, and at the community level. There is a need for immediate relief measures that should be adapted to assist the socioeconomically weaker, who may be left behind.
7. Current management approaches for COVID-19
Avoiding transmission should be the primary objective of COVID-19 treatment, particularly in those with moderate symptoms, given the uncertainty surrounding the effectiveness of currently available antiviral medications. Individuals receiving at-home care must be closely monitored, and if their health worsens, therapy must be escalated right away. Studies on the advantages of corticosteroids, weighing anti-inflammatory effects with possible hazards of viral replication, have shown conflicting findings (104). Corticosteroids may, however, be taken into consideration in situations when there are other signs, such as severe chronic obstructive pulmonary disease. Inhalers are used over nebulized medicines, which produce aerosols, to reduce the danger of airborne viral dissemination (105). Non-steroidal anti-inflammatory drugs (NSAIDs) have generated controversy die to their ability to affect epithelial cell ACE2 receptor levels and perhaps worsen viral infection (106). The specific effects of NSAID usage in COVID-19 remain uncertain. Some suggest that NSAIDs may elevate the risk of developing acute respiratory distress syndrome (ARDS) by triggering leukotriene release and bronchoconstriction (107). However, the application of NSAIDs for symptom management should be tailored to each individual. Presently, the European Medicines Agency (EMA) and the WHO do not advise against the use of NSAIDs (108). In hospital settings, acetaminophen is often preferred over NSAIDs to minimize the risk of bleeding and kidney damage (109).
Controversy has arisen regarding the use of ARBs and ACE inhibitors in COVID-19. Nonetheless, the American Society of Cardiology and the European Society of Cardiology presently do not recommend initiating or discontinuing these drugs (110). The selection of antiviral and anti-inflammatory therapies should be personalized according to each the condition of each patient, guided by infectious disease experts, and conducted within the context of a clinical trial or registry. Oxygen therapy, encompassing methods, such as nasal cannula and high-flow oxygen, is often beneficial for individuals with mild to severe COVID-19(111). Non-invasive and invasive mechanical ventilation are commonly required in situations of acute respiratory failure. Positive airway pressure is an aerosol-generating treatment; hence healthcare professionals have and must use a greater degree of personal protective equipment (112). Unless there are particular contraindications, pharmaceutical prophylaxis is used for these events and should be made available to hospitalized patients with COVID-19 due to the elevated risk of venous thromboembolism.
The PREDICT initiative by the US Agency for International Development (USAID) has significantly improved the local workforce and laboratories to identify emerging zoonotic virus risks. Investment in spillover prevention in priority countries remains a focus for USAID, with ongoing projects aimed at building a ‘One Health Workforce’ in Asia and Africa and creating interventions to halt spillover at critical high-risk human-animal interfaces (113).
8. Preventing COVID-19: Progress in vaccine advancements
Research has been conducted to evaluate a range of innovative and repurposed medicines in the battle against COVID-19(114). Among these programs, vaccinations hold promise since they could stop the spread of illness to a larger population. Before they may be used widely, the safety and efficacy of these immunizations must first be properly confirmed. It is impossible to overstate the importance of this stage since subpar immunizations run the danger of doing more damage than good via mechanisms including antibody-dependent augmentation. Therefore, meticulous testing and verification are crucial before the widespread adoption of any COVID-19 immunizations.
Technological approaches employed in the development of COVID-19 vaccines
Numerous technologies are employed by scientists and researchers globally in their endeavors to create a secure and efficient vaccine for SARS-CoV-2. Among these technologies, gene vaccines, inactivated vaccines, viral vector vaccines, and protein subunit vaccines stand out as the most promising candidates (115).
Vaccines based on protein subunits
Protein subunit vaccines, frequently administered through sophisticated systems, such as liposomes, virosomes, or polymeric nanoparticles, harness components of the pathogen to stimulate the immune system of the host (116). Liposomes and virosomes, serve as effective adjuvants and carriers for antigens and are commonly employed in the development of vaccines against SARS-CoV-2(117). For instance, research has reflected upon a cationic liposome protein subunit vaccine that incorporates the S1 component of the SARS-CoV-2 virus. This vaccine also includes two adjuvants: Monophosphoryl lipid A (MPLA), acting as a TLR4 and TLR9 agonist and CpG DNA (118). The inclusion of cationic elements such as 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) enhances the interaction of the liposome with antigen-presenting cells (115). This liposome vaccine demonstrated improved T-cell immunity, activating CD4+ and CD8+ cells and promoting IgA synthesis for potential mucosal defense (119).
Virosomes, lipid vesicles containing viral proteins, are preferred over liposomes as adjuvants due to their ability to shield pharmaceutically active compounds from degradation in endosomes until they reach the cytoplasm (120). Virosomes have previously been utilized in the delivery of vaccines for SARS-CoV and MERS-CoV. The Centre for Vaccine Development at Texas Children's Hospital, Baylor College of Medicine, is working on a subunit vaccine against SARS-CoV-2. This vaccine employs a recombinant S protein RBD, likely combined with alum or glucopyranosyl lipid A (GLA), a synthetic TLR4 agonist (121).
The Australia University of Queensland and Novavax collaborated on the development of an immunogenic virus-like nanoparticle vaccine, NVX-CoV2373, currently in phase 3 trials (NCT04611802; https://fdaaa.trialstracker.net/trial/NCT04611802/). This vaccine incorporates a recombinant S protein, demonstrating minimal reactogenicity and eliciting a T-helper 1 response without severe side-effects in the majority of individuals (122). Clover Biopharmaceuticals is also working on a highly pure S-trimer vaccine using their Trimer-Tag technology, previously employed in subunit vaccines for HIV, respiratory syncytial virus and influenza. In collaboration with GlaxoSmithKline (GSK) and Dynavax Technologies, Clover Biopharmaceuticals has completed enrollment in a phase 1 study (NCT04487210), employing the CpG 1018 adjuvant, a TLR9 agonist known to activate CD4+ and CD8+ T-cells with a favorable safety profile (123).
Vaccines with inactivated viruses
Weakened bacterial or viral pathogens used in inactivated vaccinations stimulate the immune system without actually infecting the recipient. Although these vaccinations do not provide lifelong protection, booster injections are often required to provide a long-term shielding effect. Large numbers of viral particles are propagated, condensed, and then rendered inactive using chemical and/or physical techniques to make inactivated viral vaccines. Various techniques, such as the application of ascorbic acid, binary ethylenimine, gamma irradiation and high-temperature treatment, are commonly employed to render viral particles inactive (124). The efficacy of these approaches relies on ensuring complete deactivation of the specific virus. The Wuhan Institute of Biological Products, affiliated with the China National Pharmaceutical Group (Sinopharm), actively worked on one of the initial inactivated COVID-19 vaccines (125). In the development of this vaccine, the virus undergoes growth in the Vero cell line, followed by inactivation using formalin or β-propiolactone, with alum incorporated as an adjuvant (126). All participants in the phase 1/2 clinical trials developed antibodies in response to the vaccination, with few negative side-effects (125).
The most typical adverse effects, such as discomfort at the injection site and fever, were modest and self-limiting. Phase 3 studies are currently being conducted to assess the effectiveness and long-term safety of the vaccine. Sinovac Biotech Ltd. in China is involved in developing CoronaVac (formerly PiCoVacc), another inactivated vaccine. This vaccination places genetic stability first, using the SARS-CoV-2 CN2 strain that was isolated from bronchoalveolar lavage fluid samples of hospitalized patients. The vaccine is presently in the midst of phase 3 clinical trials (NCT04456595; https://covid19.trackvaccines.org/vaccines/7/), involving a participant pool of 8,870 individuals (127). In a distinct development, the University of Wisconsin, Madison, USA, has collaborated with vaccine companies FluGen and Bharat Biotech to create an inactivated vaccine named CoroFlu, designed for intranasal delivery. Derived from FluGen's M2SR influenza vaccine, CoroFlu leverages the immune response targeting influenza. The M2SR vaccine has been adapted to incorporate the S protein gene sequences of SARS-CoV-2, to elicit an immune response against the virus (128,129). This non-invasive nasal immunization approach exhibits potential in eliciting robust mucosal and systemic immune responses to combat respiratory virus infections, providing an alternative to traditional invasive parenteral vaccination methods.
Adenovirus-based COVID-19 vaccines
Adenoviruses, with their icosahedral capsid and double-stranded linear DNA, are essential for initiating both innate and adaptive immunity in mammals. By increasing cytotoxic T-lymphocytes and releasing pro-inflammatory cytokines, they aid in the immune response. These lymphocytes are in charge of identifying and getting rid of virus-infected cells (130). Building on this method, adenoviral vectors have been extensively employed to combat a variety of illnesses, including influenza, Ebola, SARS, HIV, and recently COVID-19(131). Renowned academic institutions and pharmaceutical companies including the Jenner Institute at Oxford University, CanSino Biologics, and Johnson & Johnson have led the development of COVID-19 vaccines utilizing adenoviral vectors (132). Phase 2 clinical trials for CanSino Biologics' Ad5-nCoV vaccine are presently underway (NCT04526990; NCT04540419), and the results are promising (133). This vaccine carries the genetic code for the S protein of the SARS-CoV-2 virus and employs the non-replicating chimpanzee adenoviral vaccine vector, AZD1222. Noteworthy is its suitability for vulnerable populations, such as children, the elderly and individuals with pre-existing medical conditions, as it necessitates only a single dose and triggers a substantial immune response without causing illness (134). AstraZeneca and the University of Oxford have conducted phase 1 and phase 2 studies on AZD1222, demonstrating a promising safety profile and the successful generation of neutralizing antibodies against SARS-CoV-2 (135,136).
Adenoviral vectors are still in the early stages of development and have not yet been approved for use in the treatment of infectious diseases in humans, even though they exhibit great promise for COVID-19 vaccines. Concerns have been raised about possible inflammatory responses, as reported in AstraZeneca studies. Additionally, it is probable that individuals already have some amount of resistance to adenoviral vectors owing to their frequent exposure to them. While research and clinical trials continue, the scientific community is dedicated to developing safe and effective medicines to combat the COVID-19 pandemic and long COVID.
Nucleic acid-based vaccines
DNA vaccines or mRNA vaccines promise to be more effective than conventional immunizations. The direct administration of DNA plasmids that encode particular target antigens results in potent B- and T-cell responses with increased safety (137). These vaccinations are safe for those with impaired immune systems since they do not include any infectious organisms. Synthetic DNA vaccines facilitate the development process by enabling scalable manufacture, rapid design and preclinical testing of several candidates, and simpler regulatory approval for clinical use. Their stability at different temperatures also guarantees a longer shelf life. Currently, a gene-based vaccine is being developed that specifically targets the S protein of SARS-CoV-2. The vaccine candidate from Inovio Pharmaceuticals uses DNA-plasmid pGX9501, which was developed using MERS-CoV vaccine constructions from the past. The vaccine is currently in phase 2 clinical trials (NCT04447781; NCT04336410). It is administered intradermally and then electroporated (138). Gene vaccines also use mRNA, which operates in the cytoplasm without having to cross the nuclear membrane, in addition to DNA. mRNA vaccines are less dose-intensive than DNA vaccinations and produce strong immune system memory. However, they are less stable due to their heat lability and susceptibility to hydrolysis by circulating ribonucleases (139). This is addressed by the formulation of mRNA vaccines as lipid nanoparticles, which improve stability and host distribution. Examples include the SARS-CoV-2-targeting drugs mRNA-1273 from Moderna and BNT162b1 from Pfizer, both of which are in advanced clinical moderation (140). Despite the advancements, there are still difficulties in the global production, distribution and administration of COVID-19 vaccines.
Drugs approved for the treatment of COVID-19
Ongoing extensive clinical trials are underway to evaluate the potential effectiveness of several medications in the treatment of patients with COVID-19. The selection of these medications is based on the hypothesis that they may hinder the virus from entering the host and replicating. Various compounds, including some that have undergone human clinical trials, are currently under assessment in clinical trials as potential COVID-19 treatments. Researchers are investigating the ability of experimental drugs to impede the entry of the virus into the host and subsequent replication. While certain medications have been previously employed in treating SAR-CoV infections, others are being utilized for the first time in the context of SARS-CoV-2 infections (Table II).
Remdesivir, developed by Gilead Sciences, has received FDA approval for the treatment of patients ≥12 of age with COVID-19 requiring hospitalization. Remdesivir functions by inhibiting the RNA-dependent RNA polymerase, disrupting its interaction with the RNA of SARS-CoV-2 and thereby halting further replication (141). In a previous study, after receiving remdesivir intravenously, 36 out of 53 patients with COVID-19 exhibited an improvement, indicating positive clinical outcomes (142). Although lopinavir and ritonavir-based antiretroviral therapy have been investigated, it has not been proven to be any more effective than standard care. Umifenovir, which has been licensed for influenza prevention in China and Russia, is used for the treatment of COVID-19 due to its potential to inhibit the S protein/ACE2 interaction (143).
Research indicates that favipiravir, which inhibits RNA polymerase and is approved for use against influenza in Japan, leads to an improved clinical outcome in mild cases of COVID-19 than umifenovir (144). In small-scale clinical trials conducted in China, chloroquine has demonstrated potential in attenuating the progression of pneumonia and viral replication in patients with COVID-19 (145,146). For patients with COVID-19, the combined use of statins and ARBs holds promise for the prevention of ARDS (147). Ongoing studies are exploring the potential benefits of these combined treatments in managing the severe consequences of the illness (Fig. 5). The strategy of employing existing, approved drugs for COVID-19 treatment capitalizes on the current pharmacopeia to swiftly address the urgent global health crisis. This tactic comprises repurposing well-known pharmaceuticals that were first authorized for a range of medical conditions to target particular aspects of the SARS-CoV-2 virus or the host immune system. To make the most of these medications pharmacokinetically successful, it is important to understand the mechanisms of action and to keep in mind the safety profiles of the drug.
Remdesivir, initially developed for Ebola, has been repurposed as an antiviral for COVID-19. Its mechanism of action involves inhibiting the viral RNA polymerase, thereby disrupting viral replication (148). Lopinavir/ritonavir, FDA-approved for the treatment of HIV, is being explored for its ability to inhibit the 3CLpro enzyme in SARS-CoV-2, disrupting viral replication (149). Agents with anti-inflammatory and immunomodulatory properties, such as dexamethasone, a potent corticosteroid with strong anti-inflammatory effects, are being repurposed to alleviate the severe inflammatory responses observed in patients critically ill with COVID-19, potentially reducing mortality rates (150). The anti-inflammatory characteristics of azithromycin, an antibiotic, are currently under investigation for their ability to regulate the immune system and mitigate inflammation in individuals with COVID-19(151). Monoclonal antibodies, designed to specifically target SARS-CoV-2, are hypothesized to neutralize the virus, offering targeted therapeutic intervention.
Convalescent plasma, derived from individuals who have successfully recovered from COVID-19, contains antibodies that may neutralize the virus in infected patients, thereby enhancing the host's immune response (152). Antibiotics and antiparasitic agents, including ivermectin, known for their well-established safety profile, are undergoing examination for potential antiviral effects against SARS-CoV-2. Additionally, azithromycin, an antibiotic, is explored for its potential synergy with other treatments in COVID-19 cases (153).
9. Challenges and future prospects
One major challenge is the ongoing emergence of new SARS-CoV-2 variants. These variants may acquire increased transmissibility, be resistant to immunity from previous infections or vaccines, and may lead to more severe disease. Monitoring and adapting to these variants will be an ongoing challenge. Ensuring equitable and efficient distribution of COVID-19 vaccines in itself remains one of the major challenges. Disparities in access to vaccines can exacerbate the global health crisis and hinder efforts to achieve herd immunity. Vaccine hesitancy and misinformation continue to impede vaccination efforts. Promoting vaccine education and addressing concerns is crucial to achieving widespread vaccination and ending the pandemic. The long-term health effects of COVID-19, also referred to as ‘long COVID’, are still difficult to understand. Some individuals experience persistent symptoms and complications long after recovering from the acute phase of the disease (154,155). Healthcare systems in numerous regions around the globe are still grappling with the strain of the pandemic. Treating severe cases of COVID-19 can overwhelm hospitals and lead to delays in providing care for other serious medical conditions. The pandemic has caused severe economic and social unrest, Global cooperation and coordination are necessary to combat the pandemic effectively.
Research and the development of booster shots and updated vaccines will likely continue to address emerging variants and provide longer-lasting immunity. The development of effective antiviral drugs to treat COVID-19 may improve outcomes for those infected and reduce the severity of the disease. Achieving herd immunity through vaccination remains a key goal for ending the pandemic. Encouraging vaccination in underserved communities and improving vaccine access are essential components of this effort (156). The experience with COVID-19 underscores the need for improved pandemic preparedness, early warning systems, and global response mechanisms to mitigate the impact of future infectious disease outbreaks. The pandemic has accelerated the adoption of telemedicine and digital healthcare solutions (157). These innovations may continue to transform healthcare delivery and improve access to medical care. Addressing the mental health challenges arising from the pandemic will be a long-term prospect. Investing in mental health services and support systems is crucial for recovery. Promoting good hygiene habits and raising public health awareness can be very effective in stopping the transmission of contagious illnesses, such as COVID-19.
Numerous outbreaks of zoonotic origin have been reported over the past century, most of which have presented with severe illnesses and fatalities. One example is the aforementioned SARS-CoV-2, the cause of COVID-19. Apart from the aforementioned aspects, it is equally important to understand the preparedness beyond COVID-19 and the implications of evolving zoonotic spillovers. Vaccines are pivotal for reducing pandemic, and post-pandemic consequences. The development of vaccines requires in-depth research, large investments, and an integrated and coordinated team of diverse scientists and clinicians. Viral targets can be located and identified for their potential outbreaks and candidates for vaccine design before any adverse event. Another possibility is to lessen the emergence, by vaccinating wild animals. This will reduce zoonotic spillovers, virus evolution and amplified transmission. For example, the study by Keusch et al (158) reported that ‘vaccinating poultry or swine for emerging animal influenza A strains or camels for MERS-CoV in endemic countries’. In addition, identifying and vaccinating potential hosts may reduce the threat in endemic countries. Creating awareness among the masses, clarifying misinformation, and providing recent research updates to health workers at every level in the developed and developing world will prepare them for COVID-19 and beyond (158).
Pandemic prevention can also be managed by reducing the spillover risk by monitoring live markets, wildlife and livestock trading. The expansion of these activities is associated with the socio-economic status of the country. Some practices are a part of traditional cultural events, and hence need to be tackled mildly and judiciously. Still, these activities require strict surveillance as they are the hotspots of infectious diseases. At times trading activities link the geographical distributions, further spreading the disease and connecting and increasing the hotspots. The policy changes introduced should be evidence-based and collaborative efforts and partnerships and should be economically compatible and likely for the benefit of mankind (159,160).
Socioeconomic status, public opinion, outreach activities and communication strategies play a crucial role in preventing and preparing for COVID-19, mainly due to political polarization, variable public opinion and the associated risk specifically in the low economic strata (161). The study by AlShurman et al (162) reported that fake news associated with exaggerated risk can increase susceptibility to misinformation, and make vaccination less accepted among the masses. Parents were reported to be hesitant to regarding the vaccination of their children. Older individuals were also of the belief that vaccination may enhance their morbid status. The same study also mentioned that with proper counseling and knowledge, individuals from different beliefs and low socioeconomic distribution agreed to get vaccinated, although the number remained lower than expected (162).
Along with the importance of identifying the intersection between COVID-19 and socio-economic factors and taking appropriate measures on them, it is equally important to spread knowledge about the alternative methods or cutting-edge diagnostic approaches that are being developed or currently in use. RT-PCR is the primary method for detecting infection during the acute phase of COVID-19. The WHO and the National Health Commission of China have given the case definitions for COVID-19, which include a positive serology test even when RT-PCR is negative. The RT-PCR amplification is usually completed in a short timeframe; still the protocol includes, extraction, running time, sample processing and data management that in total requires up to 48 h. A large sample testing area where samples are transported requires even more time. RT-PCR is the gold standard for microbiological confirmation of COVID-19. Some antigen-based rapid kits are arising, such as: i) The novel Coronavirus (2019-nCov) Antigen Detection kit; ii) CLMSRDL, Sichuan Mass Spectrometry Biotechnology Co., Ltd.; iii) DIAQUICK COVID-19 Ag Cassette. These could provide rapid, point-of-care antigen tests for diagnosis of SARS-CoV-2 infection (163,164).
Infectious diseases are tackled differently, employing different approaches depending on the geographical distribution. To curb the infections some common legal measures are adopted. These measures primarily focus on the view that the need for protection is natural and necessary. Such interventions include compulsory vaccination, screening, examination, isolation and quarantines. Some studies and experts present a view that making vaccination and other measures obligatory appears to violate the patient's rights. Certain researchers are of the view, specifically in the context of COVID-19, that enforcing vaccination is unethical and does not take into consideration the ethical and religious beliefs of patients. Individuals resist undergoing vaccination due to diverse reasons, such as misinformation circulated by mass media, a lack of communication between health providers and the community, and individuals not being clear about the pros and cons of vaccination protocols. The vaccine is the only hope for this lethal infectious disease; however, it continues to face a number of ethical challenges. The study by Jalilian et al (165) and others have reported that the ethical considerations of COVID-19 fall under five categories as follows: i) Autonomy and accountability; ii) the supply of vaccines specifically to socio-economically weaker places; iii) post-vaccination safety issues; iv) the use of standard vaccine; v) fairness of reporting any adverse issues (164-166).
In principle, the equitable distribution of vaccines may not be possible due to controlled approval and limited production. Requirements, population density, policies and the capacity of low/middle-income countries vary compared to the high-income countries. To overcome these hurdles, COVAX prioritized the vaccine-receiving population and included healthcare workers and the elderly in vulnerable groups (167).
10. Conclusion and limitations
In conclusion, COVID-19 presents a range of challenges; however, there is also hope for the future. Effective vaccination, treatments, global cooperation and preparedness efforts can contribute to bringing the pandemic under control and better preparing the world to respond to future health crises. The present comprehensive review provides an in-depth and enlightening examination of the COVID-19. Millions of individuals have been affected by the SARS-CoV-2 pandemic, which has created previously unheard-of challenges for global health. The present review elaborates on several aspects of the illness, starting with its zoonotic origin and moving on to person-to-person transmission and a map of its geographic distribution across continents. An key subject included herein is the clinical manifestations COVID-19, which may vary from modest respiratory symptoms to severe instances, leading to pneumonia, ARDS and multi-organ failure. By discussing the impact of the disease on different age groups and vulnerable populations, the present review highlights the need for specialist healthcare strategies to protect those who are most susceptible. The present review also discusses several COVID-19 diagnostic methods, including molecular tests, such as PCR and antigen assays, as well as serological testing for detecting antibodies. These tests are necessary for controlling illnesses, tracking down contacts and establishing containment procedures. Effective therapies mainly include repurposed drugs, immunotherapy, cellular therapy, antiviral and other drugs. The development and administration of vaccines are observed as crucial strategies for halting the pandemic and promoting herd immunity. The present review acknowledges that significant advancements have beebn made in the comprehension and management of COVID-19; however, challenges and hurdles remain for the global healthcare systems. The challenges include handling viral alterations understanding novel varieties, combating false information and vaccination resistance and preparing for impending outbreaks. The present review provides a basis for further research and information for public health activities, and scientists working at the molecular level, helping to decrease the consequences of the epidemic and prepare for any future health crises.
In the era of long-COVID-19 presented by the silent persistence of the COVID-19 pandemic, there were a large number of studies or research in progress or in the press that could not be included or mentioned in the present review. However, the authors tried to minimize this limitation by including studies close to the discussed topic. Another limitation of the present study is that there appears to be no standardized fixed formula to cover or minimize the risk factors. Every government should accordingly make judicious decisions for the benefit of mankind. The policies adopted could not be discussed herein, as they may be circumstantial. To prevent failures in policy making and implementation, a collaborative effort among the stakeholders is necessary. The COVID-19 crisis and mandatory vaccination have become controversial.
Acknowledgements
The authors gratefully acknowledge the infrastructure facilities provided by the Department of Biochemistry, Jawaharlal Nehru Medical College, under the DST (FIST & PURSE) program.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
MM was involved in the conceptualization of the study, in the curation and investigation of data from the literature, as well as in the writing and preparation of the original draft of the manuscript. KA and RA were involved in the study design, and in the writing and editing of the manuscript. RA was involved in the curation and manuscript preparation. WA was involved in study supervision, and in the curation and investigation of data from the literature. SI was involved in the writing, review and editing of the manuscript. IQT was involved in writing, reviewing and in providing literature resources. M was involved in study supervision, in the conceptualization of the study, and in the writing, review and editing of the manuscript. MIH was involved in, study supervision and designing and image preparation. MA, NU and SH were involved in the conceptualization of the study, provision of literature and software resources, study supervision, and in the writing, review and editing of the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Cheval S, Adamescu CM, Georgiadis T, Herrnegger M, Piticar A and Legates DR: Observed and potential impacts of the COVID-19 pandemic on the environment. Int J Environ Res Public Health. 17(4140)2020.PubMed/NCBI View Article : Google Scholar | |
Woolhouse ME, Adair K and Brierley L: RNA viruses: A case study of the biology of emerging infectious diseases. Microbiol Spectr. 1(10.1128/microbiolspec.OH-0001-2012)2013.PubMed/NCBI View Article : Google Scholar | |
Markov PV, Ghafari M, Beer M, Lythgoe K, Simmonds P, Stilianakis NI and Katzourakis A: The evolution of SARS-CoV-2. Nat Rev Microbiol. 21:361–379. 2023.PubMed/NCBI View Article : Google Scholar | |
Chakraborty C, Chatterjee S, Bhattacharya M, Chopra H, Bhattacharya P, Islam MA and Dhama K: The D614G mutation helps to increase the transmissibility and reduce the virulence of SARS-CoV-2 variants through natural selection. Int J Surg. 109:171–174. 2023.PubMed/NCBI View Article : Google Scholar | |
Lythgoe KA, Hall M, Ferretti L, de Cesare M, MacIntyre-Cockett G, Trebes A, Andersson M, Otecko N, Wise EL, Moore N, et al: SARS-CoV-2 within-host diversity and transmission. Science. 372(eabg0821)2021.PubMed/NCBI View Article : Google Scholar | |
Wacharapluesadee S, Tan CW, Maneeorn P, Duengkae P, Zhu F, Joyjinda Y, Kaewpom T, Chia WN, Ampoot W, Lim BL, et al: Evidence for SARS-CoV-2 related coronaviruses circulating in bats and pangolins in Southeast Asia. Nat Commun. 12(972)2021.PubMed/NCBI View Article : Google Scholar | |
She J, Jiang J, Ye L, Hu L, Bai C and Song Y: 2019 novel coronavirus of pneumonia in Wuhan, China: Emerging attack and management strategies. Clin Transl Med. 9(19)2020.PubMed/NCBI View Article : Google Scholar | |
Hu B, Guo H, Zhou P and Shi ZL: Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol. 19:141–154. 2021.PubMed/NCBI View Article : Google Scholar | |
Worobey M, Levy JI, Malpica Serrano L, Crits-Christoph A, Pekar JE, Goldstein SA, Rasmussen AL, Kraemer MUG, Newman C, Koopmans MPG, et al: The huanan seafood wholesale market in Wuhan was the early epicenter of the COVID-19 pandemic. Science. 377:951–959. 2022.PubMed/NCBI View Article : Google Scholar | |
Reis J, Le Faou A, Buguet A, Sandner G and Spencer P: Covid-19: Early cases and disease spread. Ann Glob Health. 88(83)2022.PubMed/NCBI View Article : Google Scholar | |
Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, et al: A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 382:727–733. 2020.PubMed/NCBI View Article : Google Scholar | |
Gralinski LE and Menachery VD: Return of the coronavirus: 2019-nCoV. Viruses. 12(135)2020.PubMed/NCBI View Article : Google Scholar | |
Eurosurveillance Editorial Team. Note from the editors: World Health Organization declares novel coronavirus (2019-nCoV) sixth public health emergency of international concern. Euro Surveill. 25(200131e)2020.PubMed/NCBI View Article : Google Scholar | |
Coronaviridae Study Group of the International Committee on Taxonomy of Viruses. The species severe acute respiratory syndrome-related coronavirus: Classifying 2019-nCoV and naming it SARS-CoV-2. Nat Microbiol. 5:536–544. 2020.PubMed/NCBI View Article : Google Scholar | |
Zanin M, Xiao C, Liang T, Ling S, Zhao F, Huang Z, Lin F, Lin X, Jiang Z and Wong SS: The public health response to the COVID-19 outbreak in mainland China: A narrative review. J Thorac Dis. 12:4434–4449. 2020.PubMed/NCBI View Article : Google Scholar | |
Deng SQ and Peng HJ: Characteristics of and public health responses to the coronavirus disease 2019 outbreak in China. J Clin Med. 9(575)2020.PubMed/NCBI View Article : Google Scholar | |
World Health Organization: Coronavirus disease 2019 (COVID-19). Situation report-51, 2020. Available at: https://www.who.int/docs/default-source/coronaviruse/situation-reports/20200311-sitrep-51-covid-19.pdf?sfvrsn=1ba62e57_10. | |
Deslandes A, Berti V, Tandjaoui-Lambotte Y, Alloui C, Carbonnelle E, Zahar JR, Brichler S and Cohen Y: SARS-CoV-2 was already spreading in France in late December 2019. Int J Antimicrob Agents. 55(106006)2020.PubMed/NCBI View Article : Google Scholar | |
Pal M, Berhanu G, Desalegn C and Kandi V: Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2): An update. Cureus. 12(e7423)2020.PubMed/NCBI View Article : Google Scholar | |
Van Doremalen N, Bushmaker T, Morris DH, Holbrook MG, Gamble A, Williamson BN, Tamin A, Harcourt JL, Thornburg NJ, Gerber SI, et al: Aerosol and surface stability of SARS-CoV-2 as compared with SARS-CoV-1. N Engl J Med. 382:1564–1567. 2020.PubMed/NCBI View Article : Google Scholar | |
Aboubakr HA, Sharafeldin TA and Goyal SM: Stability of SARS-CoV-2 and other coronaviruses in the environment and on common touch surfaces and the influence of climatic conditions: A review. Transbound Emerg Dis. 68:296–312. 2021.PubMed/NCBI View Article : Google Scholar | |
Kasloff SB, Leung A, Strong JE, Funk D and Cutts T: Stability of SARS-CoV-2 on critical personal protective equipment. Sci Rep. 11(984)2021.PubMed/NCBI View Article : Google Scholar | |
Lauer SA, Grantz KH, Bi Q, Jones FK, Zheng Q, Meredith HR, Azman AS, Reich NG and Lessler J: The incubation period of coronavirus disease 2019 (COVID-19) from publicly reported confirmed cases: Estimation and application. Ann Intern Med. 172:577–582. 2020.PubMed/NCBI View Article : Google Scholar | |
Wu Y, Kang L, Guo Z, Liu J, Liu M and Liang W: Incubation period of COVID-19 caused by unique SARS-CoV-2 strains: A systematic review and meta-analysis. JAMA Netw Open. 5(e2228008)2022.PubMed/NCBI View Article : Google Scholar | |
Huang SW and Wang SF: SARS-CoV-2 entry related viral and host genetic variations: Implications on COVID-19 severity, immune escape, and infectivity. Int J Mol Sci. 22(3060)2021.PubMed/NCBI View Article : Google Scholar | |
Men K, Li Y, Wang X, Zhang G, Hu J, Gao Y, Han A, Liu W and Han H: Estimate the incubation period of coronavirus 2019 (COVID-19). Comput Biol Med. 158(106794)2023.PubMed/NCBI View Article : Google Scholar | |
Batra A, Clark JR, Kang AK, Ali S, Patel TR, Shlobin NA, Hoffman SC, Lim PH, Orban ZS, Visvabharathy L, et al: Persistent viral RNA shedding of SARS-CoV-2 is associated with delirium incidence and six-month mortality in hospitalized COVID-19 patients. Geroscience. 44:1241–1254. 2022.PubMed/NCBI View Article : Google Scholar | |
Boni MF, Lemey P, Jiang X, Lam TT, Perry BW, Castoe TA, Rambaut A and Robertson DL: Evolutionary origins of the SARS-CoV-2 sarbecovirus lineage responsible for the COVID-19 pandemic. Nat Microbiol. 5:1408–1417. 2020.PubMed/NCBI View Article : Google Scholar | |
Wu F, Zhao S, Yu B, Chen YM, Wang W, Song ZG, Hu Y, Tao ZW, Tian JH, Pei YY, et al: A new coronavirus associated with human respiratory disease in China. Nature. 579:265–269. 2020.PubMed/NCBI View Article : Google Scholar | |
He B, Zhang Y, Xu L, Yang W, Yang F, Feng Y, Xia L, Zhou J, Zhen W, Feng Y, et al: Identification of diverse alphacoronaviruses and genomic characterization of a novel severe acute respiratory syndrome-like coronavirus from bats in China. J Virol. 88:7070–7082. 2014.PubMed/NCBI View Article : Google Scholar | |
Zheng J: SARS-CoV-2: An emerging coronavirus that causes a global threat. Int J Biol Sci. 16:1678–1685. 2020.PubMed/NCBI View Article : Google Scholar | |
Temmam S, Vongphayloth K, Baquero E, Munier S, Bonomi M, Regnault B, Douangboubpha B, Karami Y, Chrétien D, Sanamxay D, et al: Author Correction: Bat coronaviruses related to SARS-CoV-2 and infectious for human cells. Nature. 607(E19)2022.PubMed/NCBI View Article : Google Scholar | |
Zhou H, Chen X, Hu T, Li J, Song H, Liu Y, Wang P, Liu D, Yang J, Holmes EC, et al: A novel bat coronavirus closely related to SARS-CoV-2 contains natural insertions at the S1/S2 cleavage site of the spike protein. Curr Biol. 30:2196–2203.e3. 2020.PubMed/NCBI View Article : Google Scholar | |
Zhou H, Ji J, Chen X, Bi Y, Li J, Wang Q, Hu T, Song H, Zhao R, Chen Y, et al: Identification of novel bat coronaviruses sheds light on the evolutionary origins of SARS-CoV-2 and related viruses. Cell. 184:4380–4391. 2021.PubMed/NCBI View Article : Google Scholar | |
Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, Zhang Q, Shi X, Wang Q, Zhang L and Wang X: Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature. 581:215–220. 2020.PubMed/NCBI View Article : Google Scholar | |
Wang H, Li X, Li T, Zhang S, Wang L, Wu X and Liu J: The genetic sequence, origin, and diagnosis of SARS-CoV-2. Eur J Clin Microbiol Infect Dis. 39:1629–1635. 2020.PubMed/NCBI View Article : Google Scholar | |
Nie J, Li Q, Zhang L, Cao Y, Zhang Y, Li T, Wu J, Liu S, Zhang M, Zhao C, et al: Functional comparison of SARS-CoV-2 with closely related pangolin and bat coronaviruses. Cell Discov. 7(21)2021.PubMed/NCBI View Article : Google Scholar | |
Yi C, Sun X, Ye J, Ding L, Liu M, Yang Z, Lu X, Zhang Y, Ma L, Gu W, et al: Key residues of the receptor binding motif in the spike protein of SARS-CoV-2 that interact with ACE2 and neutralizing antibodies. Cell Mol Immunol. 17:621–630. 2020.PubMed/NCBI View Article : Google Scholar | |
Zhang S, Qiao S, Yu J, Zeng J, Shan S, Tian L, Lan J, Zhang L and Wang X: Bat and pangolin coronavirus spike glycoprotein structures provide insights into SARS-CoV-2 evolution. Nat Commun. 12(1607)2021.PubMed/NCBI View Article : Google Scholar | |
Sharun K, Dhama K, Pawde AM, Gortázar C, Tiwari R, Bonilla-Aldana DK, Rodriguez-Morales AJ, de la Fuente J, Michalak I and Attia YA: SARS-CoV-2 in animals: Potential for unknown reservoir hosts and public health implications. Vet Q. 41:181–201. 2021.PubMed/NCBI View Article : Google Scholar | |
Dhama K, Khan S, Tiwari R, Sircar S, Bhat S, Malik YS, Singh KP, Chaicumpa W, Bonilla-Aldana DK and Rodriguez-Morales AJ: Coronavirus disease 2019-COVID-19. Clin Microbiol Rev. 33:e00028–20. 2020.PubMed/NCBI View Article : Google Scholar | |
De Wit E, Van Doremalen N, Falzarano D and Munster VJ: SARS and MERS: Recent insights into emerging coronaviruses. Nat Rev Microbiol. 14:523–534. 2016.PubMed/NCBI View Article : Google Scholar | |
Cui J, Li F and Shi ZL: Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 17:181–192. 2019.PubMed/NCBI View Article : Google Scholar | |
Wan Y, Shang J, Graham R, Baric RS and Li F: Receptor recognition by the novel coronavirus from Wuhan: An analysis based on decade-long structural studies of SARS coronavirus. J Virol. 94:e00127–20. 2020.PubMed/NCBI View Article : Google Scholar | |
Banerjee A, Doxey AC, Mossman K and Irving AT: Unraveling the zoonotic origin and transmission of SARS-CoV-2. Trends Ecol Evol. 36:180–184. 2021.PubMed/NCBI View Article : Google Scholar | |
Focosi D and Maggi F: Recombination in coronaviruses, with a focus on SARS-CoV-2. Viruses. 14(1239)2022.PubMed/NCBI View Article : Google Scholar | |
Shi J, Wen Z, Zhong G, Yang H, Wang C, Huang B, Liu R, He X, Shuai L, Sun Z, et al: Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science. 368:1016–1020. 2020.PubMed/NCBI View Article : Google Scholar | |
Oreshkova N, Molenaar RJ, Vreman S, Harders F, Oude Munnink BB, Hakze-van der Honing RW, Gerhards N, Tolsma P, Bouwstra R, Sikkema RS, et al: SARS-CoV-2 infection in farmed minks, the Netherlands, April and May 2020. Euro Surveill. 25(2001005)2020.PubMed/NCBI View Article : Google Scholar | |
Sit TH, Brackman CJ, Ip SM, Tam KWS, Law PYT, To EMW, Yu VYT, Sims LD, Tsang DNC, Chu DKW, et al: Canine SARS-CoV-2 infection. Nature. 586:776–778. 2020. | |
Zhang Q, Zhang H, Gao J, Huang K, Yang Y, Hui X, He X, Li C, Gong W, Zhang Y, et al: A serological survey of SARS-CoV-2 in cats in Wuhan. Emerg Microbes Infect. 9:2013–2019. 2020.PubMed/NCBI View Article : Google Scholar | |
Fehr AR and Perlman S: Coronaviruses: An overview of their replication and pathogenesis. Methods Mol Biol. 1282:1–23. 2015.PubMed/NCBI View Article : Google Scholar | |
Mohan SV, Hemalatha M, Kopperi H, Ranjith I and Kumar AK: SARS-CoV-2 in environmental perspective: Occurrence, persistence, surveillance, inactivation and challenges. Chem Eng J. 405(126893)2021.PubMed/NCBI View Article : Google Scholar | |
Liu DX, Liang JQ and Fung TS: Human coronavirus-229E, -OC43, -NL63, and -HKU1 (Coronaviridae). Encyclopedia Virol. (428)2021. | |
Wen C, Sun L, Zhao MC, Duan SX, Wang L and Cui XW: Clinical study of human coronavirus NL63, OC43, 229E, HKU1 infections in hospitalized children from 2015 to 2020. Infect Drug Resist. 15:1093–1101. 2022.PubMed/NCBI View Article : Google Scholar | |
Rastogi M, Pandey N, Shukla A and Singh SK: SARS coronavirus 2: From genome to infectome. Respir Res. 21:318. 2020.PubMed/NCBI View Article : Google Scholar | |
Zhu Z, Lian X, Su X, Wu W, Marraro GA and Zeng Y: From SARS and MERS to COVID-19: A brief summary and comparison of severe acute respiratory infections caused by three highly pathogenic human coronaviruses. Respir Res. 21(224)2020.PubMed/NCBI View Article : Google Scholar | |
Bourgonje AR, Abdulle AE, Timens W, Hillebrands JL, Navis GJ, Gordijn SJ, Bolling MC, Dijkstra G, Voors AA, Osterhaus AD, et al: Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19). J Pathol. 251:228–248. 2020.PubMed/NCBI View Article : Google Scholar | |
Gheblawi M, Wang K, Viveiros A, Nguyen Q, Zhong JC, Turner AJ, Raizada MK, Grant MB and Oudit GY: Angiotensin-converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: Celebrating the 20th anniversary of the discovery of ACE2. Circ Res. 126:1456–1474. 2020.PubMed/NCBI View Article : Google Scholar | |
Petrosillo N, Viceconte G, Ergonul O, Ippolito G and Petersen E: COVID-19, SARS and MERS: Are they closely related? Clin Microbiol Infect. 26:729–734. 2020.PubMed/NCBI View Article : Google Scholar | |
Xiao J, Fang M, Chen Q and He B: SARS, MERS and COVID-19 among healthcare workers: A narrative review. J Infect Public Health. 13:843–848. 2020.PubMed/NCBI View Article : Google Scholar | |
Bharati S, Podder P, Mondal MR, Podder P and Kose U: A review on epidemiology, genomic characteristics, spread, and treatments of COVID-19. Data Sci COVID-19. 487–505. 2022. | |
Harvey WT, Carabelli AM, Jackson B, Gupta RK, Thomson EC, Harrison EM, Ludden C, Reeve R and Rambaut A: COVID-19 Genomics UK (COG-UK) Consortium et al. SARS-CoV-2 variants, spike mutations and immune escape. Nat Rev Microbiol. 19:409–424. 2021.PubMed/NCBI View Article : Google Scholar | |
Marjanovic S, Romanelli RJ, Ali GC, Leach B, Bonsu M, Rodriguez-Rincon D and Ling T: COVID-19 Genomics UK (COG-UK) Consortium: Final Report. Rand Health Q. 9(24)2022.PubMed/NCBI | |
Khetran SR and Mustafa R: Mutations of SARS-CoV-2 structural proteins in the alpha, beta, gamma, and delta variants: Bioinformatics analysis. JMIR Bioinform Biotechnol. 4(e43906)2023.PubMed/NCBI View Article : Google Scholar | |
European Centre for Disease Prevention and Control: Threat Assessment Brief: Rapid increase of a SARS-CoV-2 variant with multiple spike protein mutations observed in the United Kingdom. https://www.ecdc.europa.eu/sites/default/files/documents/SARS-CoV-2-variant-multiple-spike-protein-mutations-United-Kingdom.pdf. | |
Mahmoud IS, Jarrar YB, Alshaer W and Ismail S: SARS-CoV-2 entry in host cells-multiple targets for treatment and prevention. Biochimie. 175:93–98. 2020.PubMed/NCBI View Article : Google Scholar | |
Xie Y, Karki CB, Du D, Li H, Wang J, Sobitan A, Teng S, Tang Q and Li L: Spike proteins of SARS-CoV and SARS-CoV-2 utilize different mechanisms to bind with human ACE2. Front Mol Biosci. 7(591873)2020.PubMed/NCBI View Article : Google Scholar | |
Zamorano Cuervo N and Grandvaux N: ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. Elife. 9(e61390)2020.PubMed/NCBI View Article : Google Scholar | |
Wu J, Deng W, Li S and Yang X: Advances in research on ACE2 as a receptor for 2019-CoV. Cell Mol Life Sci. 78:531–544. 2021.PubMed/NCBI View Article : Google Scholar | |
Alipoor SD and Mirsaeidi M: SARS-CoV-2 cell entry beyond the ACE2 receptor. Mol Biol Rep. 49:10715–10727. 2022.PubMed/NCBI View Article : Google Scholar | |
Guo H, Li A, Dong TY, Si HR, Hu B, Li B, Zhu Y, Shi ZL and Letko M: Isolation of ACE2-dependent and -independent sarbecoviruses from Chinese horseshoe bats. J Virol. 97(e0039523)2023.PubMed/NCBI View Article : Google Scholar | |
Shereen MA, Khan S, Kazmi A, Bashir N and Siddique R: COVID-19 infection: Emergence, transmission, and characteristics of human coronaviruses. J Adv Res. 24:91–98. 2020.PubMed/NCBI View Article : Google Scholar | |
Najafi N, Davoudi A, Izadyar H, Alishahi A, Mokhtariani A, Soleimanpourian B, Tabarrayi M, Moosazadeh M, Daftarian Z and Ahangarkani F: The effect of ACE inhibitors and ARBs on outcomes in hospitalized patients with COVID-19. Ir J Med Sci. 192:1517–1523. 2023.PubMed/NCBI View Article : Google Scholar | |
Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al: SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 181:271–280.e8. 2020.PubMed/NCBI View Article : Google Scholar | |
Liu K, Tan S, Niu S, Wang J, Wu L, Sun H, Zhang Y, Pan X, Qu X, Du P, et al: Cross-species recognition of SARS-CoV-2 to bat ACE2. Proc Natl Acad Sci USA. 118(e2020216118)2021.PubMed/NCBI View Article : Google Scholar | |
Wettstein L, Kirchhoff F and Münch J: The Transmembrane Protease TMPRSS2 as a Therapeutic Target for COVID-19 Treatment. Int J Mol Sci. 23(1351)2022.PubMed/NCBI View Article : Google Scholar | |
Guang C, Phillips RD, Jiang B and Milani F: Three key proteases-angiotensin-I-converting enzyme (ACE), ACE2 and renin-within and beyond the renin-angiotensin system. Arch Cardiovasc Dis. 105:373–385. 2012.PubMed/NCBI View Article : Google Scholar | |
Kucukoglu K, Faydalı N and Bul D: What are the drugs having potential against COVID-19? Med Chem Res. 29:1935–1955. 2020.PubMed/NCBI View Article : Google Scholar | |
Lee JS, Goldstein JM, Moon JL, Herzegh O, Bagarozzi DA Jr, Oberste MS, Hughes H, Bedi K, Gerard D, Cameron B, et al: Analysis of the initial lot of the CDC 2019-novel coronavirus (2019-nCoV) real-time RT-PCR diagnostic panel. PLoS One. 16(e0260487)2021.PubMed/NCBI View Article : Google Scholar | |
Sheikhzadeh E, Eissa S, Ismail A and Zourob M: Diagnostic techniques for COVID-19 and new developments. Talanta. 220(121392)2020.PubMed/NCBI View Article : Google Scholar | |
Udugama B, Kadhiresan P, Kozlowski HN, Malekjahani A, Osborne M, Li VYC, Chen H, Mubareka S, Gubbay JB and Chan WCW: Diagnosing COVID-19: The disease and tools for detection. ACS Nano. 14:3822–3535. 2020.PubMed/NCBI View Article : Google Scholar | |
Xie X, Zhong Z, Zhao W, Zheng C, Wang F and Liu J: Chest CT for typical coronavirus disease 2019 (COVID-19) pneumonia: Relationship to negative RT-PCR testing. Radiology. 296:E41–E45. 2020.PubMed/NCBI View Article : Google Scholar | |
Kanne JP: Chest CT findings in 2019 novel coronavirus (2019-nCoV) infections from Wuhan, China: Key points for the radiologist. Radiology. 295:16–17. 2020.PubMed/NCBI View Article : Google Scholar | |
Machado BA, Hodel KV, Barbosa-Junior VG, Soares MB and Badaro R: The main molecular and serological methods for diagnosing COVID-19: An overview based on the literature. Viruses. 13(40)2020.PubMed/NCBI View Article : Google Scholar | |
Liu YX, Zhou YH, Jiang CH, Liu J and Chen DQ: Prevention, treatment and potential mechanism of herbal medicine for Corona viruses: A review. Bioengineered. 13:5480–5508. 2022.PubMed/NCBI View Article : Google Scholar | |
Shuster A, Pechalrieu D, Jackson CB, Abegg D, Choe H and Adibekian A: Clinical antiviral drug arbidol inhibits infection by SARS-CoV-2 and variants through direct binding to the spike protein. ACS Chem Biol. 16:2845–2851. 2021.PubMed/NCBI View Article : Google Scholar | |
Kang CK, Seong MW, Choi SJ, Kim TS, Choe PG, Song SH, Kim NJ, Park WB and Oh MD: In vitro activity of lopinavir/ritonavir and hydroxychloroquine against severe acute respiratory syndrome coronavirus 2 at concentrations achievable by usual doses. Korean J Intern Med. 35(728)2020.PubMed/NCBI View Article : Google Scholar | |
RECOVERY Collaborative Group. Lopinavir-ritonavir in patients admitted to hospital with COVID-19 (RECOVERY): A randomized, controlled, open-label, platform trial. Lancet. 396:1345–1352. 2020.PubMed/NCBI View Article : Google Scholar | |
Nutho B, Mahalapbutr P, Hengphasatporn K, Pattaranggoon NC, Simanon N, Shigeta Y, Hannongbua S and Rungrotmongkol T: Why are lopinavir and ritonavir effective against the newly emerged coronavirus 2019? Atomistic insights into the inhibitory mechanisms. Biochemistry. 59:1769–1779. 2020.PubMed/NCBI View Article : Google Scholar | |
Lian N, Xie H, Lin S, Huang J, Zhao J and Lin Q: Umifenovir treatment is not associated with improved outcomes in patients with coronavirus disease 2019: A retrospective study. Clin Microbiol Infect. 26:917–921. 2020.PubMed/NCBI View Article : Google Scholar | |
Li Y, Xie Z, Lin W, Cai W, Wen C, Guan Y, Mo X, Wang J, Wang Y, Peng P, et al: Efficacy and safety of lopinavir/ritonavir or arbidol in adult patients with mild/moderate COVID-19: An exploratory randomized controlled trial. Med. 1:105–113. 2020.PubMed/NCBI View Article : Google Scholar | |
Djomkam ALZ, Olwal CO, Sala TB and Paemka L: Commentary: SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Front Oncol. 10(1448)2020.PubMed/NCBI View Article : Google Scholar | |
Li K, Meyerholz DK, Bartlett JA and McCray PB Jr: The TMPRSS2 inhibitor nafamostat reduces SARS-CoV-2 pulmonary infection in mouse models of COVID-19. mBio. 12(e0097021)2021.PubMed/NCBI View Article : Google Scholar | |
Chitsike L and Duerksen-Hughes P: Keep out! SARS-CoV-2 entry inhibitors: Their role and utility as COVID-19 therapeutics. Virol J. 18(154)2021.PubMed/NCBI View Article : Google Scholar | |
Hoffmann M, Hofmann-Winkler H, Smith JC, Krüger N, Arora P, Sørensen LK, Søgaard OS, Hasselstrøm JB, Winkler M, Hempel T, et al: Camostat mesylate inhibits SARS-CoV-2 activation by TMPRSS2-related proteases and its metabolite GBPA exerts antiviral activity. EBioMedicine. 65(103255)2021.PubMed/NCBI View Article : Google Scholar | |
Tang W, Cao Z, Han M, Wang Z, Chen J, Sun W, Wu Y, Xiao W, Liu S, Chen E, et al: Hydroxychloroquine in patients with mainly mild to moderate coronavirus disease 2019: Open label, randomised controlled trial. BMJ. 369(m1849)2020.PubMed/NCBI View Article : Google Scholar | |
Gautret P, Lagier JC, Parola P, Hoang VT, Meddeb L, Mailhe M, Doudier B, Courjon J, Giordanengo V, Vieira VE, et al: Hydroxychloroquine and azithromycin as a treatment of COVID-19: Results of an open-label non-randomized clinical trial. Int J Antimicrob Agents. 56(105949)2020.PubMed/NCBI View Article : Google Scholar | |
Manivannan E, Karthikeyan C, Moorthy NSHN and Chaturvedi SC: The rise and fall of Chloroquine/Hydroxychloroquine as compassionate therapy of COVID-19. Front Pharmacol. 12(584940)2021.PubMed/NCBI View Article : Google Scholar | |
Kumar S, Dutta D, Ravichandiran V and Sukla S: Monoclonal antibodies: A remedial approach to prevent SARS-CoV-2 infection. 3 Biotech. 12(227)2022.PubMed/NCBI View Article : Google Scholar | |
Javalkote VS, Kancharla N, Bhadra B, Shukla M, Soni B, Sapre A, Goodin M, Bandyopadhyay A and Dasgupta S: CRISPR-based assays for rapid detection of SARS-CoV-2. Methods. 203:594–603. 2022.PubMed/NCBI View Article : Google Scholar | |
Joung J, Ladha A, Saito M, Segel M, Bruneau R, Huang MW, Kim NG, Yu X, Li J, Walker BD, et al: Point-of-care testing for COVID-19 using SHERLOCK diagnostics. medRxiv [Preprint]: May 8, 2020 doi: 10.1101/2020.05.04.20091231. | |
Yang SM, Lv S, Zhang W and Cui Y: Microfluidic point-of-care (POC) devices in early diagnosis: A review of opportunities and challenges. Sensors (Basel). 22(1620)2022.PubMed/NCBI View Article : Google Scholar | |
Rizvi ZA, Babele P, Madan U, Sadhu S, Tripathy MR, Goswami S, Mani S, Dikshit M and Awasthi A: Pharmacological potential of Withania somnifera (L.) Dunal and Tinospora cordifolia (Willd.) Miers on the experimental models of COVID-19, T cell differentiation, and neutrophil functions. Front Immunol. 14(1138215)2023.PubMed/NCBI View Article : Google Scholar | |
Amponsah SK, Tagoe B, Adams I and Bugyei KA: Efficacy and safety profile of corticosteroids and non-steroidal anti-inflammatory drugs in COVID-19 management: A narrative review. Front Pharmacol. 13(1063246)2022.PubMed/NCBI View Article : Google Scholar | |
Falk JA, Minai OA and Mosenifar Z: Inhaled and systemic corticosteroids in chronic obstructive pulmonary disease. Proc Am Thorac Soc. 5:506–512. 2008.PubMed/NCBI View Article : Google Scholar | |
Kushner P, McCarberg BH, Grange L, Kolosov A, Haveric AL, Zucal V, Petruschke R and Bissonnette S: The use of non-steroidal anti-inflammatory drugs (NSAIDs) in COVID-19. NPJ Prim Care Respir Med. 32(35)2022.PubMed/NCBI View Article : Google Scholar | |
Robb CT, Goepp M, Rossi AG and Yao C: Non-steroidal anti-inflammatory drugs, prostaglandins, and COVID-19. Br J Pharmacol. 177:4899–4920. 2020.PubMed/NCBI View Article : Google Scholar | |
Sokolowska M, Rovati GE, Diamant Z, Untersmayr E, Schwarze J, Lukasik Z, Sava F, Angelina A, Palomares O, Akdis CA, et al: Effects of non-steroidal anti-inflammatory drugs and other eicosanoid pathway modifiers on antiviral and allergic responses: EAACI task force on eicosanoids consensus report in times of COVID-19. Allergy. 77:2337–2354. 2022.PubMed/NCBI View Article : Google Scholar | |
Kanchanasurakit S, Arsu A, Siriplabpla W, Duangjai A and Saokaew S: Acetaminophen use and risk of renal impairment: A systematic review and meta-analysis. Kidney Res Clin Pract. 39(81)2020.PubMed/NCBI View Article : Google Scholar | |
De Spiegeleer A, Bronselaer A, Teo JT, Byttebier G, De Tré G, Belmans L, Dobson R, Wynendaele E, Van De Wiele C, Vandaele F, et al: The effects of ARBs, ACEis, and statins on clinical outcomes of COVID-19 infection among nursing home residents. J Am Med Dir Assoc. 21:909–914.e2. 2020.PubMed/NCBI View Article : Google Scholar | |
Geng S, Mei Q, Zhu C, Yang T, Yang Y, Fang X and Pan A: High flow nasal cannula is a good treatment option for COVID-19. Heart Lung. 49:444–445. 2020.PubMed/NCBI View Article : Google Scholar | |
Lockhart SL, Duggan LV, Wax RS, Saad S and Grocott HP: Personal protective equipment (PPE) for both anesthesiologists and other airway managers: Principles and practice during the COVID-19 pandemic. Can J Anaesth. 67:1005–1015. 2020.PubMed/NCBI View Article : Google Scholar | |
Gilardi KVK and Mazet JAK: The United States Agency for international development emerging pandemic threats PREDICT project-global detection of emerging wildlife viral zoonoses. Fowler's Zoo Wild Animal Med Curr Ther. 9:110–116. 2019. | |
Chakraborty C, Sharma AR, Bhattacharya M, Agoramoorthy G and Lee SS: The drug repurposing for COVID-19 clinical trials provide very effective therapeutic combinations: Lessons learned from major clinical studies. Front Pharmacol. 12(704205)2021.PubMed/NCBI View Article : Google Scholar | |
Okuyama R: mRNA and adenoviral vector vaccine platforms utilized in COVID-19 vaccines: Technologies, ecosystem, and future directions. Vaccines (Basel). 11(1737)2023.PubMed/NCBI View Article : Google Scholar | |
Wang N, Chen M and Wang T: Liposomes used as a vaccine Adjuvant-delivery system: From basics to clinical immunization. J Control Release. 303:130–150. 2019.PubMed/NCBI View Article : Google Scholar | |
Tretiakova DS and Vodovozova EL: Liposomes as adjuvants and vaccine delivery systems. Biochem (Mosc) Suppl Ser A Membr Cell Biol. 16:1–20. 2022.PubMed/NCBI View Article : Google Scholar | |
Petkar KC, Patil SM, Chavhan SS, Kaneko K, Sawant KK, Kunda NK and Saleem IY: An overview of Nanocarrier-based adjuvants for vaccine delivery. Pharmaceutics. 13(455)2021.PubMed/NCBI View Article : Google Scholar | |
Karunakaran B, Gupta R, Patel P, Salave S, Sharma A, Desai D, Benival D and Kommineni N: Emerging trends in lipid-based vaccine delivery: A special focus on developmental strategies, fabrication methods, and applications. Vaccines (Basel). 11(661)2023.PubMed/NCBI View Article : Google Scholar | |
Ali H, Akbar M, Iqbal B, Ali F, Sharma NK, Kumar N, Najmi A, Albratty M, Alhazmi HA, Madkhali OA, et al: Virosome: An engineered virus for vaccine delivery. Saudi Pharm J. 31:752–764. 2023.PubMed/NCBI View Article : Google Scholar | |
Nakanishi T, Hayashi A, Kunisawa J, Tsutsumi Y, Tanaka K, Yashiro-Ohtani Y, Nakanishi M, Fujiwara H, Hamaoka T and Mayumi T: Fusogenic liposomes efficiently deliver exogenous antigen through the cytoplasm into the MHC class I processing pathway. Eur J Immunol. 30:1740–1747. 2000.PubMed/NCBI View Article : Google Scholar | |
Marchese AM, Zhou X, Kinol J, Underwood E, Woo W, McGarry A, Beyhaghi H, Áñez G, Toback S and Dunkle LM: NVX-CoV2373 vaccine efficacy against hospitalization: A post hoc analysis of the PREVENT-19 phase 3, randomized, placebo-controlled trial. Vaccine. 41:3461–3466. 2023.PubMed/NCBI View Article : Google Scholar | |
Frederiksen LSF, Zhang Y, Foged C and Thakur A: The long road toward COVID-19 herd immunity: Vaccine platform technologies and mass immunization strategies. Front Immunol. 11(1817)2020.PubMed/NCBI View Article : Google Scholar | |
Sanders B, Koldijk M and Schuitemaker H: Inactivated viral vaccines. Vaccine Analysis: Strategies Principles Control: 45-80, 2014 doi: 10.1007/978-3-662-45024-6_2. | |
Wang C, Chen LY, Lu QB and Cui F: Vaccination with the inactivated vaccine (Sinopharm BBIBP-CorV) ensures protection against SARS-CoV-2 related disease. Vaccines (Basel). 10(920)2022.PubMed/NCBI View Article : Google Scholar | |
Hu L, Sun J, Wang Y, Tan D, Cao Z, Gao L, Guan Y, Jia X and Mao J: A review of inactivated COVID-19 vaccine development in China: Focusing on safety and efficacy in special populations. Vaccines (Basel). 11(1045)2023.PubMed/NCBI View Article : Google Scholar | |
Soysal A, Gönüllü E, Karabayır N, Alan S, Atıcı S, Yıldız İ, Engin H, Çivilibal M and Karaböcüoğlu M: Comparison of immunogenicity and reactogenicity of inactivated SARS-CoV-2 vaccine (CoronaVac) in previously SARS-CoV-2 infected and uninfected health care workers. Hum Vaccin Immunother. 17:3876–3880. 2021.PubMed/NCBI View Article : Google Scholar | |
Pagliusi S, Jarrett S, Hayman B, Kreysa U, Prasad SD, Reers M, Hong Thai P, Wu K, Zhang YT, Baek YO, et al: Emerging manufacturers engagements in the COVID-19 vaccine research, development and supply. Vaccine. 38:5418–5423. 2020.PubMed/NCBI View Article : Google Scholar | |
Thanh Le T, Andreadakis Z, Kumar A, Gómez Román R, Tollefsen S, Saville M and Mayhew S: The COVID-19 vaccine development landscape. Nat Rev Drug Discov. 19:305–306. 2020.PubMed/NCBI View Article : Google Scholar | |
Chaplin DD: Overview of the immune response. J Allergy Clin Immunol. 125 (2 Suppl 2):S3–S23. 2010.PubMed/NCBI View Article : Google Scholar | |
Chavda VP, Bezbaruah R, Valu D, Patel B, Kumar A, Prasad S, Kakoti BB, Kaushik A and Jesawadawala M: Adenoviral vector-based vaccine platform for COVID-19: Current status. Vaccines (Basel). 11(432)2023.PubMed/NCBI View Article : Google Scholar | |
Elkashif A, Alhashimi M, Sayedahmed EE, Sambhara S and Mittal SK: Adenoviral vector-based platforms for developing effective vaccines to combat respiratory viral infections. Clin Transl Immunology. 10(e1345)2021.PubMed/NCBI View Article : Google Scholar | |
Zhu FC, Guan XH, Li YH, Huang JY, Jiang T, Hou LH, Li JX, Yang BF, Wang L, Wang WJ, et al: Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 396:479–488. 2020.PubMed/NCBI View Article : Google Scholar | |
Joe CCD, Jiang J, Linke T, Li Y, Fedosyuk S, Gupta G, Berg A, Segireddy RR, Mainwaring D, Joshi A, et al: Manufacturing a chimpanzee adenovirus-vectored SARS-CoV-2 vaccine to meet global needs. Biotechnol Bioeng. 119:48–58. 2022.PubMed/NCBI View Article : Google Scholar | |
Asano M, Okada H, Itoh Y, Hirata H, Ishikawa K, Yoshida E, Matsui A, Kelly EJ, Shoemaker K, Olsson U and Vekemans J: Immunogenicity and safety of AZD1222 (ChAdOx1 nCoV-19) against SARS-CoV-2 in Japan: A double-blind, randomized controlled phase 1/2 trial. Int J Infect Dis. 114:165–174. 2022.PubMed/NCBI View Article : Google Scholar | |
AstraZeneca: AZD1222 US Phase III primary analysis confirms safety and efficacy. AstraZeneca, 2021. Available at: https://www.astrazeneca.com/content/astraz/media-centre/press-releases/2021/azd1222-us-phase-iii-primary-analysis-confirms-safety-and-efficacy.html. | |
Khan KH: DNA vaccines: Roles against diseases. Germs. 3:26–35. 2013.PubMed/NCBI View Article : Google Scholar | |
Zhao G, Zhang Z, Ding Y, Hou J, Liu Y, Zhang M, Sui C, Wang L, Xu X, Gao X and Kou Z: A DNA vaccine encoding the full-length spike protein of beta variant (B.1.351) elicited broader cross-reactive immune responses against other SARS-CoV-2 variants. Vaccines (Basel). 11(513)2023.PubMed/NCBI View Article : Google Scholar | |
Chavda VP, Jogi G, Dave S, Patel BM, Vineela Nalla L and Koradia K: mRNA-Based vaccine for COVID-19: They are new but not unknown! Vaccines (Basel). 11(507)2023.PubMed/NCBI View Article : Google Scholar | |
Hussain A, Yang H, Zhang M, Liu Q, Alotaibi G, Irfan M, He H, Chang J, Liang XJ, Weng Y and Huang Y: mRNA vaccines for COVID-19 and diverse diseases. J Control Release. 345:314–333. 2022.PubMed/NCBI View Article : Google Scholar | |
Gordon CJ, Tchesnokov EP, Woolner E, Perry JK, Feng JY, Porter DP and Götte M: Remdesivir is a direct-acting antiviral that inhibits RNA-dependent RNA polymerase from severe acute respiratory syndrome coronavirus 2 with high potency. J Biol Chem. 295:6785–6797. 2020.PubMed/NCBI View Article : Google Scholar | |
Grein J, Ohmagari N, Shin D, Diaz G, Asperges E, Castagna A, Feldt T, Green G, Green ML and Lescure FX: Compassionate Use of Remdesivir for Patients with Severe Covid-19. N Engl J Med. 382:2327–2336. 2020.PubMed/NCBI View Article : Google Scholar | |
Chen C, Zhang Y, Huang J, Yin P, Cheng Z, Wu J, Chen S, Zhang Y, Chen B, Lu M, et al: Favipiravir versus arbidol for clinical recovery rate in moderate and severe adult COVID-19 Patients: A prospective, multicenter, open-label, randomized controlled clinical trial. Front Pharmacol. 12(683296)2021.PubMed/NCBI View Article : Google Scholar | |
Joshi S, Parkar J, Ansari A, Vora A, Talwar D, Tiwaskar M, Patil S and Barkate H: Role of favipiravir in the treatment of COVID-19. Int J Infect Dis. 102:501–508. 2021.PubMed/NCBI View Article : Google Scholar | |
Lei ZN, Wu ZX, Dong S, Yang DH, Zhang L, Ke Z, Zou C and Chen ZS: Chloroquine and hydroxychloroquine in the treatment of malaria and repurposing in treating COVID-19. Pharmacol Ther. 216(107672)2020.PubMed/NCBI View Article : Google Scholar | |
Mallat J, Hamed F, Balkis M, Mohamed MA, Mooty M, Malik A, Nusair A and Bonilla MF: Hydroxychloroquine is associated with slower viral clearance in clinical COVID-19 patients with mild to moderate disease. Medicine (Baltimore). 99(e23720)2020.PubMed/NCBI View Article : Google Scholar | |
Yang G, Tan Z, Zhou L, Yang M, Peng L, Liu J, Cai J, Yang R, Han J, Huang Y and He S: Effects of Angiotensin II receptor blockers and ACE (Angiotensin-Converting Enzyme) inhibitors on virus infection, inflammatory status, and clinical outcomes in patients with COVID-19 and hypertension: A single-center retrospective study. Hypertension. 76:51–58. 2020.PubMed/NCBI View Article : Google Scholar | |
Eastman RT, Roth JS, Brimacombe KR, Simeonov A, Shen M, Patnaik S and Hall MD: Remdesivir: A review of its discovery and development leading to emergency use authorization for treatment of COVID-19. ACS Cent Sci. 6:672–683. 2020.PubMed/NCBI View Article : Google Scholar | |
Uzunova K, Filipova E, Pavlova V and Vekov T: Insights into antiviral mechanisms of remdesivir, lopinavir/ritonavir and chloroquine/hydroxychloroquine affecting the new SARS-CoV-2. Biomed Pharmacother. 131(110668)2020.PubMed/NCBI View Article : Google Scholar | |
Bahsoun A, Fakih Y, Zareef R, Bitar F and Arabi M: Corticosteroids in COVID-19: Pros and cons. Front Med (Lausanne). 10(1202504)2023.PubMed/NCBI View Article : Google Scholar | |
Venditto VJ, Haydar D, Abdel-Latif A, Gensel JC, Anstead MI, Pitts MG, Creameans J, Kopper TJ, Peng C and Feola DJ: Immunomodulatory effects of azithromycin revisited: Potential applications to COVID-19. Front Immunol. 12(574425)2021.PubMed/NCBI View Article : Google Scholar | |
Peng HT, Rhind SG and Beckett A: Convalescent plasma for the prevention and treatment of COVID-19: A systematic review and quantitative analysis. JMIR Public Health Surveill. 7(e25500)2021.PubMed/NCBI View Article : Google Scholar | |
Pandey A, Nikam AN, Shreya AB, Mutalik SP, Gopalan D, Kulkarni S, Padya BS, Fernandes G, Mutalik S and Prassl R: Potential therapeutic targets for combating SARS-CoV-2: Drug repurposing, clinical trials and recent advancements. Life Sci. 256(117883)2020.PubMed/NCBI View Article : Google Scholar | |
Aiyegbusi OL, Hughes SE, Turner G, Rivera SC, McMullan C, Chandan JS, Haroon S, Price G, Davies EH, Nirantharakumar K, et al: Symptoms, complications and management of long COVID: A review. J R Soc Med. 114:428–442. 2021.PubMed/NCBI View Article : Google Scholar | |
Hosseinzadeh P, Zareipour M, Baljani E and Moradali MR: Social consequences of the COVID-19 pandemic. A systematic review. Invest Educ Enferm. 40(e10)2022.PubMed/NCBI View Article : Google Scholar | |
Wouters OJ, Shadlen KC, Salcher-Konrad M, Pollard AJ, Larson HJ, Teerawattananon Y and Jit M: Challenges in ensuring global access to COVID-19 vaccines: Production, affordability, allocation, and deployment. Lancet. 397:1023–1034. 2021.PubMed/NCBI View Article : Google Scholar | |
McNeely JA: Nature and COVID-19: The pandemic, the environment, and the way ahead. Ambio. 50:767–781. 2021.PubMed/NCBI View Article : Google Scholar | |
Keusch GT, Amuasi JH, Anderson DE, Daszak P, Eckerle I, Field H, Koopmans M, Lam SK, Das Neves CG, Peiris M, et al: Pandemic origins and a One Health approach to preparedness and prevention: Solutions based on SARS-CoV-2 and other RNA viruses. Proc Natl Acad Sci USA. 119(e2202871119)2022.PubMed/NCBI View Article : Google Scholar | |
Morens DM, Taubenberger JK and Fauci AS: Universal coronavirus vaccines-an urgent need. N Engl J Med. 386:297–299. 2022.PubMed/NCBI View Article : Google Scholar | |
Loh EH, Zambrana-Torrelio C, Olival KJ, Bogich TL, Johnson CK, Mazet JA, Karesh W and Daszak P: Targeting transmission pathways for emerging zoonotic disease surveillance and control. Vector Borne Zoonotic Dis. 15:432–437. 2015.PubMed/NCBI View Article : Google Scholar | |
Pike J, Bogich T, Elwood S, Finnoff DC and Daszak P: Economic optimization of a global strategy to address the pandemic threat. Proc Natl Acad Sci USA. 111:18519–18523. 2014.PubMed/NCBI View Article : Google Scholar | |
AlShurman BA, Khan AF, Mac C, Majeed M and Butt ZA: What demographic, social, and contextual factors influence the intention to use COVID-19 vaccines: A scoping review. Int J Environ Res Public Health. 18(9342)2021.PubMed/NCBI View Article : Google Scholar | |
Dinnes J, Sharma P, Berhane S, van Wyk SS, Nyaaba N, Domen J, Taylor M, Cunningham J, Davenport C, Dittrich S, et al: Rapid, point-of-care antigen tests for diagnosis of SARS-CoV-2 infection. Cochrane Database Syst Rev. 7(CD013705)2022.PubMed/NCBI View Article : Google Scholar | |
Malekzadeh R, Abedi G, Ziapour A, Yıldırım M and Amirkhanlou A: Analysis of ethical considerations of COVID-19 vaccination: Lessons for future. BMC Med Ethics. 24(91)2023.PubMed/NCBI View Article : Google Scholar | |
Jalilian H, Amraei M, Javanshir E, Jamebozorgi K and Faraji-Khiavi F: Ethical considerations of the vaccine development process and vaccination: A scoping review. BMC Health Serv Res. 23(255)2023.PubMed/NCBI View Article : Google Scholar | |
Chung JY, Thone MN and Kwon YJ: COVID-19 vaccines: The status and perspectives in delivery points of view. Adv Drug Deliv Rev. 170:1–25. 2021.PubMed/NCBI View Article : Google Scholar | |
Forman R, Shah S, Jeurissen P, Jit M and Mossialos E: COVID-19 vaccine challenges: What have we learned so far and what remains to be done? Health Policy. 125:553–567. 2021.PubMed/NCBI View Article : Google Scholar |