ABSTRACT
The ongoing COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has spurred unprecedented research efforts worldwide. This comprehensive scientific review synthesizes key research developments and understandings, providing a nuanced exploration of the dynamic landscape of COVID-19. The virological section examines structural variations, viral replication mechanisms, immune responses and the impact of emerging variants of concern. Transmission dynamics are scrutinized, with a focus on airborne transmission, super-spreading events and the often elusive asymptomatic and pre-symptomatic transmission phases. Clinical manifestations and severity are elucidated, exploring genetic factors, immunopathology and long-term sequelae. Advancements in diagnostics are discussed, highlighting molecular techniques, rapid antigen tests and serological assays, while therapeutic developments encompass antiviral agents, immunomodulatory treatments and the challenges in drug development. Vaccine research is scrutinized, with in-depth analyses of mRNA and vector-based vaccines, including efficacy against variants and distribution challenges. The socioeconomic impact section evaluates economic repercussions, health disparities and long-term societal changes. The review underscores the importance of ongoing research, emphasizing unanswered questions and knowledge gaps. Future research directions explore novel technologies and collaborative efforts for improved diagnostics, treatments and vaccines. In conclusion, this review provides a panoramic view of the current state of COVID-19 research, offering insights for future pandemic preparedness and global health strategies.
INTRODUCTION
The emergence of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) marked a pivotal moment in global health, leading to the ongoing COVID-19 pandemic. SARS-CoV-2 is a member of the coronavirus family, sharing genetic similarities with other coronaviruses such as SARS-CoV and MERS-CoV.1–3 Comprehensive genomic analyses have traced the evolutionary history of SARS-CoV-2, revealing a zoonotic origin likely involving a spillover event from wildlife to humans. These investigations have identified potential reservoirs and intermediate hosts, shedding light on the complex dynamics of the virus’s adaptation to human hosts. The zoonotic transmission dynamics of SARS-CoV-2 have been a subject of intense scrutiny.4,5 Investigations into wildlife markets and the identification of coronaviruses in bats, pangolins and other animals have provided valuable insights into the origins of the virus. The intricate interplay between wildlife, domestic animals and human populations underscores the multifaceted nature of zoonotic transmission, with genetic analyses aiding in the reconstruction of transmission pathways.
Structural variations in the SARS-CoV-2 genome have been a focus of research to understand the virus’s adaptability and infectivity.6 Notably, mutations in the spike protein, particularly in the receptor-binding domain, have been implicated in the virus’s ability to bind to human cells.7,8 Structural analyses of the virus’s components, including the spike protein and viral envelope, contribute to the comprehension of its pathogenicity and the development of potential therapeutic interventions. The viral replication mechanisms of SARS-CoV-2 are complex and involve intricate interactions between viral and host factors. Investigations into the molecular machinery of viral replication have identified key host cellular components that facilitate or inhibit the virus’s ability to replicate efficiently.9 Adaptive changes in the viral genome, driven by selective pressures, contribute to the virus’s ability to evade host immune responses and potentially alter its transmissibility. The immune response dynamics to SARS-CoV-2 have been a critical area of research, encompassing T-cell and B-cell responses as well as the role of antibodies in immunity.10,11 Understanding the immune response is pivotal for vaccine development and therapeutic strategies. The virus’s ability to evade the host immune system through various mechanisms, including mutations in key epitopes, has significant implications for the ongoing development of effective vaccines and treatments. The emergence of variants of concern has added a layer of complexity to the dynamics of SARS-CoV-2.12,13 These variants, characterized by specific mutations in the viral genome, have been associated with altered transmissibility, immune escape and potential impacts on the efficacy of existing vaccines. Surveillance efforts aimed at monitoring the emergence and spread of variants are crucial for adapting public health strategies and vaccine development to the evolving nature of the virus. The significance of understanding key research developments in the context of SARS-CoV-2 and COVID-19 cannot be overstated, as it is paramount for effective public health strategies, therapeutic interventions and vaccine development. Comprehensive knowledge of the virus’s molecular characteristics, transmission dynamics, clinical manifestations and host-immune interactions is critical for mitigating the impact of the pandemic and preparing for future infectious disease threats. The first crucial aspect lies in deciphering the molecular intricacies of SARS-CoV-2. Understanding the genetic evolution and structural variations of the virus provides essential insights into its origin, adaptability and pathogenicity.14 This information is fundamental for developing targeted therapeutics and vaccines that can specifically address the virus’s vulnerabilities while minimizing the potential for resistance.
Key research developments are also essential in unraveling the transmission dynamics of SARS-CoV-2.15 Insight into modes of transmission, such as airborne, surface contamination and the roles of asymptomatic carriers, informs the design and implementation of public health interventions. This knowledge guides the development of evidence-based strategies for effective containment, contact tracing and quarantine measures, thereby limiting the spread of the virus within communities. Understanding the clinical manifestations and severity of COVID-19 is crucial for optimizing patient care and resource allocation within healthcare systems.16 Identifying risk factors for severe outcomes, elucidating the immunopathology underlying severe cases and recognizing the long-term effects of the disease contribute to the development of tailored treatment protocols and rehabilitation strategies for those recovering from COVID-19. The significance of key research developments extends into the realm of diagnostics, where ongoing advancements shape testing strategies and accuracy. Comprehensive knowledge of the strengths and limitations of diagnostic tools, including RT-PCR, rapid antigen tests and serological assays, is essential for effective case identification, contact tracing and monitoring the progression of the pandemic.17 Research developments also drive therapeutic advancements, providing insights into potential antiviral agents, immunomodulatory treatments and supportive care strategies. A deep understanding of the virus’s replication mechanisms and immune evasion strategies is critical for developing targeted therapeutics that can disrupt viral replication and modulate the host immune response.18 Researchers have examined the relationship between the virus biology and the cellular response to infection.19
In the context of vaccines, ongoing research developments play a central role in the design, testing and optimization of COVID-19 vaccines vaccination is still the most effective and safest way to avoid this disease.20,21 Knowledge of the immunogenicity, efficacy and safety profiles of different vaccine platforms inform vaccination strategies, including the timing of booster doses and addressing emerging variants of concern. The socioeconomic impact of the pandemic underscores the need for research developments that elucidate the economic, social and psychological consequences of COVID-19.22 This knowledge informs policy decisions, resource allocation and the development of support systems for vulnerable populations. The impact of the SARS-CoV-2 virus on global public health has been profound, necessitating a comprehensive understanding of the repercussions and the recalibration of research priorities to address immediate challenges and prepare for future pandemics.23 The rapid spread of COVID-19 highlighted the interconnectedness of the global community and the imperative for collaborative, data-driven responses to emerging infectious diseases. The immediate impact on global public health was manifested in overwhelmed healthcare systems, shortages of medical supplies and the strain on healthcare professionals. The unprecedented demand for testing, treatments and hospital resources underscored the need for robust public health infrastructures that can rapidly adapt to emerging threats. Research priorities were swiftly redirected to address critical gaps in knowledge regarding transmission dynamics, clinical management and public health interventions to curb the spread of the virus.
One of the key lessons learned from the global response to the pandemic is the importance of early detection and surveillance.24 Research efforts focused on developing and optimizing diagnostic tools, with an emphasis on speed, accuracy and scalability. The ability to quickly identify and isolate cases, trace contacts and implement quarantine measures emerged as essential components of effective pandemic control. Understanding the impact of COVID-19 on vulnerable populations and healthcare disparities became a research priority.25 Disparities in healthcare access, outcomes and socioeconomic factors were amplified during the pandemic. Research efforts aimed at unraveling the root causes of these disparities were initiated to inform public health policies that address the structural inequities contributing to differential health outcomes. The pandemic catalyzed a paradigm shift in vaccine development and distribution. Research priorities pivoted towards the rapid development of safe and effective
COVID-19 vaccines, utilizing innovative platforms such as mRNA technology and adenovirus vectors. Global collaborations and initiatives, such as COVAX, were established to ensure equitable access to vaccines, emphasizing the interconnectedness of global health and the importance of addressing the pandemic on a global scale.26
The long-term consequences of COVID-19 on mental health emerged as an additional research priority. Prolonged periods of social isolation, economic uncertainty and the direct impact of the virus on neurological systems highlighted the need for comprehensive research into mental health outcomes and the development of support systems for that affected.27 Furthermore, the pandemic exposed gaps in our preparedness for emerging infectious diseases. Research priorities shifted towards the development of proactive surveillance systems, early warning mechanisms and strategies for rapid deployment of resources in anticipation of future outbreaks. The importance of a One Health approach, integrating human, animal and environmental health, gained prominence as a means of preventing and mitigating future pandemics.28 The impact of synthesizing crucial research findings in the context of SARS-CoV-2 has been instrumental in shaping our understanding of the virus and guiding public health responses. The systematic compilation of research outcomes, spanning virology, transmission dynamics, clinical manifestations and therapeutic interventions, has facilitated a more nuanced comprehension of COVID-19. This synthesis has enabled the identification of patterns, correlations and critical factors influencing disease outcomes, paving the way for evidence-based decision-making in the global response to the pandemic. However, this synthesis has also highlighted significant gaps and challenges in our current knowledge of SARS-CoV-2. The intricate interactions between the virus and the host immune system, the determinants of viral transmissibility and the long-term effects of COVID-19 are areas where our understanding remains incomplete. Gaps in knowledge regarding the efficacy and safety of therapeutic interventions, the dynamics of viral evolution and the factors contributing to severe disease outcomes pose challenges for the development of targeted treatments and preventive measures.
Evaluation of gaps and challenges in current knowledge has underscored the need for ongoing research efforts to address critical unanswered questions. The evolving nature of the pandemic, coupled with the emergence of new variants of concern.29 emphasizes the necessity for continuous surveillance and research to adapt public health strategies and vaccine development. Gaps in understanding the immune response to SARS-CoV-2, particularly in diverse populations, highlight the importance of inclusive and representative research designs to ensure the effectiveness of interventions across different demographic groups.30 Challenges in diagnostics, including the variability in test performance and the impact of emerging variants on test accuracy, demand ongoing research to refine testing strategies. Therapeutic challenges, such as the identification of optimal treatment regimens, addressing drug resistance concerns and managing potential long-term sequelae, necessitate a concerted research effort. The complexity of the virus-host interaction and the factors influencing transmission dynamics also demand continuous investigation to inform preventive measures and public health interventions.
Virology of SARS-CoV-2
Structural variations and implications
Receptor Binding Domain (RBD) alterations in the spike protein of SARS-CoV-2 represent a critical aspect of the virus’s evolution and its interaction with host cells.31 The RBD is a specific region within the spike protein responsible for binding to the Angiotensin-Converting Enzyme 2 (ACE2) receptor on the surface of human cells, initiating the process of viral entry. As such, alterations in the RBD can have profound implications for viral infectivity, transmissibility and the virus’s ability to evade host immune responses. Structural analyses have identified various alterations in the RBD, with mutations affecting key amino acid residues involved in the interaction with the ACE2 receptor.32 These alterations can result in changes to the binding affinity between the spike protein and the ACE2 receptor, influencing the efficiency of viral entry into host cells. Certain RBD mutations have been associated with increased transmissibility, potentially enhancing the virus’s ability to establish infection in the human population.
The spike protein of the SARS-CoV-2 virus has garnered significant scientific attention due to its crucial role in mediating viral entry into host cells and its potential impact on viral infectivity and immune evasion.33 Spike protein mutations have been a focal point of research as they can influence the virus’s ability to bind to ACE2 receptor on human cells, a key step in the viral entry process. Structural analyses of the spike protein have revealed various mutations, particularly within the RBD, the region that directly interacts with the ACE2 receptor.34 These mutations can result in alterations to the protein’s conformation, potentially enhancing or diminishing its affinity for the ACE2 receptor. Such changes may influence the efficiency of viral entry into host cells, affecting transmissibility and infectivity. One study has found that mutations on RBD of omicron variant resulted in stronger binding to human ACE2 receptor.35 Several spike protein mutations have been identified as variants of concern, such as those found in the Alpha, Beta, Gamma and Delta variants.36 These variants exhibit distinctive patterns of spike protein mutations, which have been associated with increased transmissibility and, in some cases, altered disease severity. The Delta variant, for instance, harbors mutations in the RBD that have been linked to enhanced transmissibility compared to earlier variants.37 Importantly, spike protein mutations can impact the efficacy of neutralizing antibodies generated through natural infection or vaccination.38 Some mutations within the spike protein may result in a reduced recognition of the virus by neutralizing antibodies, potentially compromising the protective immune response.39 This phenomenon has led to concerns about the potential for immune escape, where certain variants may partially evade the immunity generated by prior infection or vaccination. The dynamic nature of spike protein mutations necessitates ongoing genomic surveillance to monitor the emergence and spread of new variants. Research efforts focus on characterizing the functional consequences of these mutations, including their impact on viral entry, replication and interactions with the host immune system. This knowledge is crucial for adapting diagnostic strategies, optimizing vaccine formulations and developing targeted therapeutics that account for the evolving viral landscape. Receptor binding domain alterations can also play a role in immune evasion.40 The RBD is a primary target for neutralizing antibodies generated in response to natural infection or vaccination. Changes in the RBD may impact the recognition and binding of neutralizing antibodies, potentially reducing the efficacy of immune responses. This phenomenon raises concerns about the potential for certain variants with RBD alterations to partially escape immunity, influencing vaccine effectiveness and the course of the pandemic. Ongoing research focuses on characterizing the functional consequences of RBD alterations, including their impact on viral entry kinetics, the ability to establish infection in different cell types and the potential for increased pathogenicity. Understanding these alterations at the molecular level is crucial for the development of diagnostics, vaccines and therapeutic interventions that can effectively target and mitigate the impact of SARS-CoV-2 variants.
Host-SARS-CoV-2 interactions
Host-virus interactions form the foundation of the complex relationship between SARS-CoV-2 and the human host. These interactions involve a series of intricate molecular and cellular events that determine the course of infection, the host’s immune response and the outcome of the disease. Understanding the nuances of these interactions is critical for deciphering the pathogenesis of COVID-19 and devising targeted therapeutic interventions. The coronavirus virion contains essential structural proteins, including Spike (S), Envelope (E), Membrane (M), Nucleocapsid (N) and sometimes haemagglutinin-esterase.41 The viral genome, a positive-sense, single-stranded RNA (+ssRNA), is enveloped by the N protein, while the M and E proteins facilitate its incorporation into the viral particle during assembly. The initial step in host-virus interactions is the attachment of the virus to host cells. SARS-CoV-2 primarily gains entry into human cells by binding its spike protein to ACE2 receptor on the surface of respiratory epithelial cells. This specific interaction is central to the virus’s tropism for the respiratory tract and subsequent respiratory symptoms associated with COVID-19.
The S proteins protrude from the viral envelope and play a crucial role in binding to cellular receptors, such as ACE2, promoting viral entry and fusion with the host cell membrane. Following attachment, host-virus interactions involve viral entry and replication. The virus enters the host cell through a process known as endocytosis, where the viral genetic material is released into the cell’s cytoplasm. Once inside, the virus hijacks the host cellular machinery to replicate its genome and produce viral proteins. This interplay between viral and host factors dictates the efficiency of viral replication and the severity of the infection. Upon entry, the genomic RNA is released and undergoes immediate translation of ORF1a and ORF1b, resulting in polyproteins pp1a and pp1ab, which are further processed into non-structural proteins (nsps). These nsps form the viral replication and transcription complex, leading to the formation of viral replication organelles, including Double-Membrane Vesicles (DMVs), Convoluted Membranes (CMs) and small open Double-Membrane Spherules (DMSs), facilitating viral RNA replication and transcription of subgenomic mRNAs (sg mRNAs). The translated structural proteins are transported to the Endoplasmic Reticulum (ER) membranes, where they interact with newly produced genomic RNA encapsulated by N protein, leading to budding into secretory vesicular compartments within the ER-to-Golgi Intermediate Compartment (ERGIC). Finally, virions are secreted from the infected cell via exocytosis.41
SARS-CoV-2 employs rapid protein mutation to evade immune responses, notably altering the spike protein; a key target for neutralizing antibodies.42 This enables the virus to reduce antibody effectiveness from prior immunity. Moreover, the virus suppresses specific immune cell function, like T lymphocytes and natural killer cells, prolonging viral shedding and potentially worsening disease outcomes. The virus has evolved various mechanisms to avoid detection and neutralization by the host’s immune defenses, allowing for its successful replication and persistence within the human host. Understanding these immune evasion strategies is crucial for informing the development of effective vaccines, therapeutic interventions and public health measures. The virus also employs strategies to evade the host’s innate immune response, which serves as the first line of defense against viral infections. SARS-CoV-2 can inhibit the production of type I interferons, signaling proteins crucial for coordinating the antiviral response.43 By suppressing the early innate immune response, the virus can gain a foothold in the host and establish infection before more robust adaptive immune responses are activated. Antigenic variation, another immune evasion strategy, involves the alteration of viral antigens, to escape recognition by pre-existing immunity.44 This mechanism contributes to the phenomenon of viral variants with enhanced transmissibility and the potential to partially evade immunity generated by previous infections or vaccinations. Understanding these immune evasion strategies informs ongoing efforts to develop effective countermeasures against SARS-CoV-2. Vaccine design considers the evolving viral landscape, aiming to elicit immune responses that can recognize and neutralize a broad range of viral variants. Therapeutic interventions also take into account the virus’s ability to modulate the host immune response, targeting specific viral proteins or pathways to enhance antiviral immunity.
The host’s immune response plays a pivotal role in shaping the trajectory of host-virus interactions.45 Upon infection, the innate immune system is activated, leading to the release of signaling molecules such as interferons that induce an antiviral state in neighboring cells. This early immune response aims to restrict viral spread and provide time for the adaptive immune system to mount a specific response against the virus. In the context of host-virus interactions, the adaptive immune response is characterized by the activation of T cells and B cells. T cells, specifically cytotoxic T cells, target and eliminate infected cells, while B cells produce antibodies that recognize and neutralize the virus. The interplay between viral antigens and host immune cells determines the efficacy of the adaptive immune response, influencing the resolution of infection and the development of immunological memory.
However, SARS-CoV-2 has evolved strategies to evade the host immune response. This includes the aforementioned mutation of key viral proteins, such as the spike protein, to escape neutralization by antibodies. The virus can also modulate immune cell function, impairing the activation of T cells and suppressing the overall immune response. These immune evasion strategies contribute to the virus’s ability to establish persistent infections and, in some cases, lead to severe disease outcomes. Host-virus interactions extend beyond the respiratory system, with evidence of the virus affecting multiple organ systems. The cardiovascular, gastrointestinal and central nervous systems can be impacted, resulting in a diverse range of clinical manifestations observed in COVID-19 patients.46 The mechanisms underlying these systemic effects are complex and involve a combination of direct viral invasion, immune responses and inflammatory processes.
Variants of Concern and their impact on transmissibility and immune evasion
Variants of Concern (VOCs) in the context of SARS-CoV-2 represent strains of the virus that have undergone significant genetic changes, raising concerns due to their potential impact on transmissibility, disease severity and immune evasion. The emergence of VOCs underscores the dynamic nature of the virus and the need for continuous surveillance to monitor genetic variations.47 Several VOCs have been identified, including the Alpha, Beta, Gamma and Delta variants, each associated with unique sets of mutations that can influence viral properties (Table 1). One major aspect of concern is the impact of variants on transmissibility.48 Certain mutations within the spike protein and other genomic regions of the virus can enhance its ability to infect host cells and spread within populations. The Delta variant, for example, has been associated with increased transmissibility compared to earlier strains, contributing to its global prevalence. Understanding the genetic basis for heightened transmissibility is crucial for predicting and mitigating the spread of these variants. Immune evasion is another significant concern associated with VOCs.42 Mutations in the spike protein or other viral components may alter the virus’s antigenic profile, potentially reducing the recognition and neutralization by antibodies generated through natural infection or vaccination. This phenomenon raises concerns about the effectiveness of existing immunity against certain variants. The Beta variant, for instance, has exhibited resistance to neutralization by some monoclonal antibodies and a potential reduction in vaccine efficacy, highlighting the importance of ongoing research to assess the impact of variants on immune responses. The evolution of VOCs is shaped by selective pressures and the emergence of certain mutations may confer advantages to the virus in terms of host interactions.49 Variants with increased transmissibility and immune evasion may have a fitness advantage, allowing them to outcompete other strains in the population. Continuous genomic surveillance, coupled with laboratory studies to assess the functional consequences of mutations, is crucial for understanding the dynamics of VOCs and guiding public health responses.
Variant Name | Key Mutations | Origin | Transmission Rate | Vaccine Efficacy | Clinical Severity | Public Health Impact |
---|---|---|---|---|---|---|
Alpha (B.1.1.7) | N501Y, P681H, Δ69/70 | United Kingdom | Higher | Reduced | Similar/mild | Increased transmissibility, early vaccine escape. |
Beta (B.1.351) | E484K, K417N, N501Y | South Africa | Higher | Reduced | Similar/mild | Reduced vaccine efficacy, potential reinfections. |
Gamma (P.1) | E484K, K417T, N501Y | Brazil | Higher | Reduced | Similar/mild | Reduced vaccine efficacy, potential reinfections. |
Delta (B.1.617.2) | L452R, P681R, Δ157/158, Δ478K | India | Higher | Reduced | Possibly severe | Increased transmissibility, potential vaccine escape. |
Omicron (B.1.1.529) | Numerous mutations | South Africa/ Botswana | Very high | Not yet determined | Not yet determined | Potential for immune evasion, under investigation. |
Epsilon (B.1.427/B.1.429) | L452R, D614G | United States | Higher | Reduced | Similar/mild | Increased transmissibility, potential vaccine escape. |
Mu (B.1.621) | E484K, N501Y | Colombia | Higher | Reduced | Similar/mild | Under investigation, potential vaccine escape. |
Lambda (C.37) | L452Q, F490S | Peru | Higher | Reduced | Similar/mild | Increased transmissibility, potential vaccine escape. |
Transmission dynamics
Airborne transmission dynamics
Aerosolization of respiratory droplets is a critical factor influencing the transmission dynamics of respiratory viruses, including SARS-CoV-2.50 Respiratory droplets are typically generated during activities such as talking, coughing, sneezing, or breathing and can vary in size. Larger droplets tend to settle quickly, while smaller droplets, known as aerosols, can remain suspended in the air for extended periods. The aerodynamic behavior of respiratory droplets depends on their size and the surrounding environmental conditions.51 Larger droplets, with diameters greater than 5 micrometers, are subject to gravity and tend to fall to the ground within a short distance from the source. However, smaller droplets, especially those less than 5 micrometers in diameter, can linger in the air and may travel over longer distances. These aerosols can be inhaled by individuals in close proximity to the source or even in enclosed spaces where ventilation is limited.
The aerosolization of respiratory droplets is particularly relevant to the transmission of respiratory viruses in indoor environments. Poor ventilation, crowded spaces and prolonged exposure increase the risk of inhaling infectious aerosols. SARS-CoV-2 has been shown to be present in respiratory aerosols, especially in individuals with symptomatic and asymptomatic infections, highlighting the potential for airborne transmission. This mode of transmission is particularly concerning as it poses challenges for traditional infection control measures that primarily target larger respiratory droplets. Understanding the dynamics of aerosolized respiratory droplets is crucial for implementing effective public health measures.52 Improved ventilation, air filtration systems and the use of air purifiers can help reduce the concentration of infectious aerosols in indoor settings. The wearing of masks, especially those with filtration capabilities, can also mitigate the emission and inhalation of respiratory droplets. Maintaining physical distancing remains important, particularly in enclosed spaces, to reduce the risk of close-range exposure to aerosols.
Environmental factors play a significant role in influencing the transmission dynamics of respiratory viruses, including SARS-CoV-2.53 The survival and spread of respiratory viruses are influenced by various environmental conditions that affect viral stability, transmission routes and host susceptibility. Understanding these factors is crucial for implementing effective public health measures to control the spread of infectious agents. Temperature and humidity are key environmental factors that influence the transmission of respiratory viruses.54 Cold and dry conditions are generally associated with increased respiratory virus transmission. In such environments, respiratory droplets containing viral particles may remain viable for longer periods and the dry air can compromise the mucous membranes in the respiratory tract, potentially enhancing viral entry and infection. These conditions contribute to the seasonality observed in the transmission of respiratory viruses, with winter months often associated with increased viral activity. Ventilation is another critical environmental factor influencing transmission.55 Inadequate ventilation in indoor spaces can lead to the accumulation of respiratory droplets and aerosols, increasing the risk of exposure to infectious particles. Well-ventilated areas, on the other hand, help disperse airborne particles, reducing the concentration of viral agents in the air. Proper ventilation, combined with air filtration systems, is essential for minimizing the risk of airborne transmission, particularly in enclosed spaces where individuals may spend extended periods.
Ultraviolet (UV) radiation from sunlight has been shown to have a detrimental effect on the viability of respiratory viruses.56 UV radiation can damage the genetic material of viruses, reducing their infectivity. This environmental factor contributes to the decreased stability of respiratory viruses in outdoor settings, where exposure to sunlight is more direct. Outdoor environments generally pose a lower risk of transmission compared to confined indoor spaces, emphasizing the importance of outdoor activities in reducing the spread of respiratory viruses. Population density and human behavior are additional environmental factors influencing transmission dynamics.57,58 Crowded settings with close interpersonal contact increase the risk of virus transmission, especially in the absence of proper preventive measures. Human behavior, such as mask-wearing, hand hygiene and adherence to physical distancing guidelines, significantly impacts the effectiveness of control measures in mitigating the spread of respiratory viruses. Environmental factors also extend to the presence of fomites, surfaces contaminated with infectious particles.59 The stability of respiratory viruses on surfaces varies depending on the material, temperature and humidity. Regular cleaning and disinfection of frequently touched surfaces, combined with proper hand hygiene, are crucial for interrupting the chain of transmission through fomites.
Super spreading events
Super-spreading events, characterized by the disproportionate transmission of infectious agents by a small number of individuals, have played a significant role in the dynamics of the COVID-19 pandemic caused by SARS-CoV-2.60 These events are marked by the transmission of the virus to a larger number of people than would be expected based on the average transmission patterns. Understanding the characteristics and contributing factors of super-spreading events is crucial for tailoring effective public health interventions. Several characteristics define super-spreading events. First, these events often occur in specific settings, such as indoor gatherings, where close interpersonal contact is prevalent. Second, certain individuals, known as super-spreaders, play a disproportionate role in transmission. These individuals may shed a higher viral load or engage in behaviors that facilitate widespread transmission, contributing significantly to the overall spread of the virus. Third, the timing of exposure and viral shedding plays a role, as individuals may be more infectious during certain stages of the infection.
Contributing factors to super-spreading events include the presence of high viral loads in certain individuals, conducive environmental conditions and behavioral factors. Super-spreaders, who may have higher viral loads, can amplify transmission when they come into contact with a large number of susceptible individuals.61 Settings with poor ventilation, where respiratory droplets and aerosols can accumulate, contribute to the efficient spread of the virus. Additionally, behaviors such as singing, shouting and prolonged close contact increase the risk of transmission during these events. Contact tracing and testing strategies become particularly important in the aftermath of super-spreading events.62 Rapid identification and isolation of cases and their contacts help break the chain of transmission and prevent further spread. Public health messaging plays a role in raising awareness about the risk factors and behaviors associated with super-spreading events, encouraging individuals to adopt preventive measures.
Asymptomatic and pre-symptomatic transmission
Asymptomatic and pre-symptomatic transmission of SARS-CoV-2, the virus responsible for COVID-19, has been a significant challenge in controlling the spread of the virus.63 Individuals who are infected with the virus but do not show symptoms (asymptomatic) or have not yet developed symptoms (pre-symptomatic) can unknowingly transmit the virus to others. This phenomenon has profound implications for public health strategies, as it complicates efforts to identify and isolate cases based solely on symptomatology. Asymptomatic individuals are those who are infected with the virus but do not develop noticeable symptoms throughout the course of their infection. They may carry a viral load similar to symptomatic individuals, making them capable of transmitting the virus to others. The challenge with asymptomatic transmission lies in the difficulty of identifying and isolating these individuals without widespread testing. Studies have suggested that a significant proportion of SARS-CoV-2 infections may be asymptomatic, contributing to the silent spread of the virus within communities.64,65 Pre-symptomatic individuals, on the other hand, are those who have been infected with the virus but have not yet developed symptoms.66 During this pre-symptomatic period, which can precede the onset of symptoms by a few days, individuals may unknowingly spread the virus to others. This phase highlights the importance of identifying and isolating cases before symptoms manifest. However, the variability in the incubation period and the potential for a substantial proportion of transmission occurring during the pre-symptomatic phase pose challenges for timely interventions.67
The dynamics of asymptomatic and pre-symptomatic transmission underscore the importance of widespread testing, contact tracing and isolation measures in controlling the spread of SARS-CoV-2. Diagnostic testing, particularly in settings with high transmission rates, allows for the identification of individuals who may be asymptomatic or pre-symptomatic carriers.68 Early detection enables timely isolation and reduces the risk of further transmission, contributing to the overall containment of the virus. Public health interventions, such as mask-wearing, physical distancing and hygiene practices, play a crucial role in mitigating the impact of asymptomatic and pre-symptomatic transmission. These measures are designed to reduce the risk of respiratory droplet and aerosol transmission, which is a key mode of spread for SARS-CoV-2. The implementation of these preventive measures, combined with vaccination efforts, aims to break the chains of transmission and protect vulnerable populations.
Diagnostic advances
Molecular diagnostics
Molecular diagnostics, particularly reverse transcription Polymerase Chain Reaction (RT-PCR), has been a cornerstone in the detection and diagnosis of SARS-CoV-2, the virus responsible for COVID-19.69 RT-PCR is a highly sensitive and specific technique that amplifies viral RNA to detect the presence of the virus. It involves the conversion of viral RNA into complementary DNA (cDNA) using reverse transcriptase, followed by PCR amplification and fluorescence-based detection. This method allows for the accurate identification of SARS-CoV-2 even in individuals with low viral loads, providing a reliable tool for diagnosing active infections. Despite its widespread use and effectiveness, RT-PCR has some limitations. The technique requires specialized laboratory equipment and trained personnel, making it less suitable for point-of-care testing. Additionally, the turnaround time for RT-PCR results can vary, ranging from a few hours to a day, which may impact timely decision-making in certain situations.70 False-negative results can occur, especially if the sample is collected during the early or late stages of infection when viral loads are lower.71 Sensitivity can also be influenced by the quality of the sample collection and the presence of inhibitors in the sample.
Next-Generation Sequencing (NGS) has emerged as a powerful tool for understanding the genomic landscape of SARS-CoV-2 and detecting emerging variants.72 NGS allows for the high-throughput sequencing of the entire viral genome, providing a comprehensive view of the genetic diversity within viral populations. This capability is crucial for surveillance efforts, as it enables the detection of new variants, monitoring of mutations associated with changes in transmissibility or immune evasion and assessment of the virus’s evolutionary dynamics over time. NGS has been particularly instrumental in identifying variants of concern, such as the Alpha, Beta, Gamma and Delta variants of SARS-CoV-2.73 By analyzing the genetic makeup of the virus, researchers can track the emergence and spread of specific variants and assess their potential impact on public health. The ability of NGS to detect mutations in the spike protein, receptor-binding domain and other key regions helps in understanding the potential implications for diagnostics, therapeutics and vaccine effectiveness. Despite its advantages, NGS also presents challenges, including the need for sophisticated laboratory infrastructure, bioinformatics expertise for data analysis and longer turnaround times compared to traditional diagnostic methods.74 The high cost associated with NGS may limit its widespread use for routine diagnostic purposes, making it more suitable for research, surveillance and monitoring of viral evolution.
Rapid antigen tests and their role
Rapid antigen tests have played a crucial role in the global response to the COVID-19 pandemic, providing a rapid and accessible method for detecting the presence of SARS-CoV-2, the virus responsible for COVID-19.75,76 These tests operate on the principle of detecting viral proteins, specifically the presence of viral antigens, within a patient’s sample. Unlike molecular diagnostic methods such as RT-PCR, rapid antigen tests do not require sophisticated laboratory infrastructure and can deliver results within minutes at the point of care.
One key consideration in evaluating rapid antigen tests is their sensitivity and specificity. Sensitivity refers to the test’s ability to correctly identify individuals with an active infection, while specificity indicates the test’s ability to correctly identify individuals without the infection. Rapid antigen tests generally exhibit high specificity, meaning they are effective in correctly identifying individuals who are not infected.77 However, their sensitivity can vary and they may be less accurate in detecting infections, particularly in individuals with lower viral loads or during the early stages of infection. It is essential to consider the trade-off between speed and sensitivity when deploying rapid antigen tests in different settings.
Despite potential limitations in sensitivity, rapid antigen tests have found widespread use in various settings, including point-of-care applications.78 The rapid turnaround time makes them valuable tools for quickly identifying and isolating infectious individuals, especially in high-risk environments such as healthcare facilities, airports, schools and community testing centres. Point-of-care testing reduces the time between sample collection and result reporting, enabling prompt decision-making and public health interventions.79 The accessibility of rapid antigen tests contributes to broader testing coverage and supports efforts to control the spread of the virus. Point-of-care applications also extend to decentralized and remote settings where access to centralized laboratories may be limited. Mobile testing units, pharmacies and community clinics can deploy rapid antigen tests to facilitate on-the-spot testing, providing timely information to individuals and public health authorities.80 The ease of use and minimal equipment requirements make rapid antigen tests suitable for a variety of testing scenarios, enhancing the overall testing capacity and surveillance efforts. It is crucial to recognize that the role of rapid antigen tests complements, rather than replaces, more sensitive molecular diagnostic methods like RT-PCR. While rapid antigen tests are effective in quickly identifying infectious individuals, negative results may require confirmation through molecular testing, especially in high-risk or symptomatic individuals. The integration of both rapid antigen tests and molecular methods contributes to a comprehensive testing strategy that addresses the different needs of diverse populations.
Thus, rapid antigen tests have played a pivotal role in the fight against the COVID-19 pandemic, providing a rapid and accessible means of detecting SARS-CoV-2 at the point of care. Their high specificity and quick turnaround time make them valuable tools for various applications, including point-of-care testing in diverse settings. As part of a comprehensive testing strategy, rapid antigen tests contribute to broader testing coverage, aiding in the identification and isolation of infectious individuals to mitigate the spread of the virus.
Serological assays and antibody kinetics
Serological assays, which detect antibodies in blood samples, have been integral in understanding the immune response to SARS-CoV-2. Antibody kinetics, the patterns of antibody production and persistence over time, has been a focus of research to unravel the dynamics of the immune response following infection or vaccination. A recent investigation utilizing high-throughput neutralization and serology assays uncovered correlated yet notably diverse humoral immune responses in a substantial cohort of individuals infected with SARS-CoV-2.81 Longitudinal studies examining antibody responses contribute valuable insights into the durability of immunity and the potential for reinfection.
Following SARS-CoV-2 infection or vaccination, the immune system produces antibodies, including Immunoglobulin G (IgG), against viral proteins, such as the spike protein. The kinetics of antibody responses involves an initial rise, peak and subsequent decline, forming distinct phases in the immune response. Longitudinal studies have revealed that the magnitude and duration of antibody responses can vary among individuals.82 Factors such as the severity of the initial infection, age and underlying health conditions influence the strength and persistence of antibody responses. Longitudinal studies on antibody responses have shown that while antibody levels may decline over time, especially after the initial peak, a substantial proportion of individuals maintain detectable antibodies for months.83,84 The decline in antibody levels does not necessarily indicate a loss of immunity; instead, it may reflect the transition from the acute phase of the immune response to a more sustained and memory-driven phase. Memory B cells and T cells, critical components of the adaptive immune system, contribute to long-term immune memory and play a role in responding to future encounters with the virus.85
Implications for immunity and reinfection stem from the understanding of antibody kinetics and the broader immune response. Longitudinal studies suggest that while antibody levels may wane, the immune system retains the ability to mount a rapid and effective response upon re-exposure to the virus.85,86 This phenomenon indicates the presence of immunological memory, which is a key component of protective immunity. The durability of immunity, both antibody-mediated and cell-mediated, is a crucial factor in assessing the risk of reinfection and the potential need for booster vaccinations. Studies on reinfection have shown that while instances of reinfection with SARS-CoV-2 are relatively rare, they can occur.87 The dynamics of reinfection are influenced by various factors, including the individual’s immune status, the presence of emerging variants and the elapsed time since the initial infection.88 Reinfections are generally associated with milder symptoms compared to primary infections, highlighting the role of pre-existing immunity in mitigating disease severity. The implications of antibody kinetics and longitudinal studies extend beyond individual immunity to inform public health strategies. Understanding the dynamics of immune responses aids in optimizing vaccination schedules, determining the need for booster doses and refining recommendations for post-infection surveillance.89 Longitudinal studies provide valuable data for modeling the population-level impact of immunity and assessing the potential for future waves of infection.
Therapeutic developments
Antiviral agents and their mechanisms
Antiviral agents represent a critical component in the therapeutic arsenal against viral infections, including SARS-CoV-2. These agents can be classified into two main categories: Direct-Acting Antivirals (DAAs) that target viral components and host-targeted therapies that interfere with host cell processes essential for viral replication.90 Remdesivir, a direct-acting antiviral, gained prominence as an early therapeutic option for COVID-19.91 It functions as a nucleotide analogue, inhibiting the viral RNA-dependent RNA polymerase and disrupting the synthesis of viral RNA. By incorporating into the viral RNA chain, Remdesivir causes premature termination of RNA synthesis, limiting the production of infectious viral particles. Clinical trials have demonstrated its efficacy in reducing the time to recovery in hospitalized COVID-19 patients, particularly those requiring supplemental oxygen. Other direct-acting antivirals targeting SARS-CoV-2 include protease inhibitors and polymerase inhibitors. Protease inhibitors, such as lopinavir/ /ritonavir, interfere with viral replication by inhibiting the activity of viral proteases crucial for processing viral polyproteins. However, their efficacy in COVID-19 has been debated and some studies have not shown significant clinical benefits. Paxlovid, a combination of nirmatrelvir and ritonavir, demonstrates significant effectiveness and safety as an antiviral treatment for SARS-CoV-2.92 Nirmatrelvir functions as a reversible covalent peptidomimetic inhibitor targeting the main protease of SARS-CoV-2, pivotal for viral replication. In tandem, ritonavir acts as a pharmacokinetic enhancer, permanently inhibiting the cytochrome CYP3A4 enzyme, which metabolizes nirmatrelvir rapidly, thus extending its half-life and availability. Furthermore,
Polymerase inhibitors, similar to Remdesivir, target the viral RNA polymerase and disrupt viral RNA synthesis.93 Favipiravir is one such inhibitor that has shown promise in reducing the duration of symptoms in mild to moderate COVID-19 cases.94,95 Host-targeted therapies represent another avenue in antiviral drug development, aiming to disrupt host cell processes essential for viral replication. One example is monoclonal antibodies that target specific viral proteins or the host cell receptors involved in viral entry. Monoclonal antibodies, such as casirivimab and imdevimab, have received emergency use authorization for the treatment of mild to moderate COVID-19 cases; reducing the risk of progression to severe disease.96–98 Other host-targeted therapies include drugs that modulate the host immune response. Corticosteroids, such as dexamethasone, have demonstrated efficacy in reducing mortality and improving outcomes in severe COVID-19 cases.99 By dampening excessive immune responses and mitigating inflammation, corticosteroids play a crucial role in managing the hyperinflammatory state associated with severe disease.100
In addition to small molecule drugs, research efforts are exploring the potential of repurposed drugs with antiviral properties.101 Drugs like ivermectin, originally developed as an antiparasitic agent, have been investigated for their potential antiviral effects against SARS-CoV-2.102 However, the efficacy and safety of these repurposed drugs are subjects of ongoing research and debate. Combination therapies, involving the simultaneous use of multiple antiviral agents with complementary mechanisms of action, are also being explored to enhance efficacy and reduce the risk of drug resistance. For instance, Favipiravir and ivermectin exhibit combined antiviral effectiveness against SARS-CoV-2 in laboratory settings. This combination is currently undergoing evaluation in a randomized controlled clinical study (NCT05155527).103 Also pairing of lopinavir/ritonavir with antibiotic azithromycin proves to be a potent medication combination for handling the COVID-19 virus or potential future viral pandemic outbreaks.104 The evolving landscape of antiviral therapeutics for COVID-19 underscores the importance of ongoing research, clinical trials and evidence-based approaches to optimize treatment strategies.
Immunomodulatory treatments
Immunomodulatory treatments, including corticosteroids and monoclonal antibodies, have played a crucial role in managing the immune response and mitigating severe outcomes in COVID-19 patients.105 As a potent anti-inflammatory medication, dexamethasone modulates the immune system by suppressing the excessive inflammatory response observed in severe cases. Clinical trials, such as the RECOVERY trial, demonstrated that dexamethasone reduced mortality in hospitalized COVID-19 patients requiring supplemental oxygen or mechanical ventilation.106,107 The success of dexamethasone highlights the importance of targeted immunomodulation in preventing the harmful effects of hyperinflammation associated with severe COVID-19.
Monoclonal antibodies represent another class of immunomodulatory treatments with the ability to neutralize SARS-CoV-2 and modulate the host immune response. Casirivimab and imdevimab, a combination of monoclonal antibodies, have received emergency use authorization for the treatment of mild to moderate COVID-19 cases.108 By binding to specific viral proteins or the virus itself, monoclonal antibodies prevent viral entry into host cells and promote the clearance of the virus from the body. These immunomodulatory effects contribute to reducing the severity and duration of symptoms in individuals with early-stage COVID-19. Monoclonal antibodies are particularly valuable for high-risk individuals who may benefit from early intervention to prevent progression to severe disease.109
The efficacy of immunomodulatory treatments extends beyond antiviral effects, encompassing their impact on the dysregulated immune response observed in severe COVID-19 cases.110 Recently, a single-cell RNA sequencing (scRNA-seq) investigation examined COVID-19 pediatric patients across various disease severities, including those with encephalopathy complications. This study unveiled new insights into immune characteristics associated with COVID-19-related encephalopathy. Notably, the findings shed light on potential therapeutic targets for managing this condition, particularly in patients with acute necrotizing encephalopathy.111 Recent studies suggest that individuals experiencing Long COVID (LC), also known as Post-Acute Sequelae of SARS-CoV-2 infection (PASC), exhibit persistent symptoms and health issues long after the acute phase of the infection has resolved.112 One aspect of LC involves dysregulation of T cells, which are a crucial component of the immune system responsible for recognizing and fighting off viral infections. Additionally, there is ongoing inflammation and an immune response that lacks coordination in its efforts to combat the SARS-CoV-2 virus. This dysregulated and prolonged immune response may contribute to the persistence of symptoms and health complications seen in individuals with LC.
In addition to dexamethasone and monoclonal antibodies, other immunomodulators are being investigated for their potential in managing the inflammatory aspects of the disease. Tocilizumab, an Interleukin-6 (IL-6) receptor antagonist, has shown promise in reducing mortality and improving outcomes in severe COVID-19 cases with elevated IL-6 levels.113 The modulation of specific immune pathways, such as the IL-6 pathway, reflects the targeted approach of immunomodulatory treatments in addressing the immune dysregulation associated with severe disease.114 A study assessed the effectiveness of the IL-1 antagonist anakinra, the IL-6 antagonist tocilizumab and corticosteroids in managing mild to moderate cases of COVID-19.115 The results demonstrated that anakinra was notably effective in reducing several biomarkers such as ferritin, D-dimer, LDH, CRP and WBCs in COVID-19 patients compared to tocilizumab. Additionally, anakinra was associated with a decrease in the number of patients requiring admission to the Intensive Care Unit (ICU). These findings suggest that clinicians may consider using anakinra in early-stage COVID-19 to reduce the need for ICU admission and mechanical ventilation.
While immunomodulatory treatments have demonstrated efficacy, their use is carefully considered based on disease severity and patient characteristics. In milder cases or during the early stages of infection, the focus may be on antiviral agents and supportive care. However, in severe cases characterized by hyperinflammation and a cytokine storm, immunomodulatory treatments become essential components of the therapeutic strategy.116 One recent study provided a comprehensive overview of single-target and multi-target drug discovery techniques, emphasizing the growing preference for multi-target approaches in combating SARS-CoV-2.117 It discusses medications in clinical trials, highlighting the shift in focus towards vaccine studies over drug therapy trials. The potential of natural compounds from plants as antiviral agents against SARS-CoV-2 is explored, but challenges remain due to limited understanding of their mechanisms of action, underscoring the need for safe and effective COVID-19 treatments with minimal side effects. The evolving landscape of COVID-19 therapeutics emphasizes the need for personalized and targeted approaches based on the patient’s clinical status and the stage of the disease.118 Combining antiviral agents with immunomodulatory treatments represents a comprehensive strategy to address both viral replication and the dysregulated immune response.
Challenges in drug development and repurposing
Drug development and repurposing, especially in the context of emerging viral infections like COVID-19, pose several challenges that impact the speed and efficacy of therapeutic interventions.119 One significant challenge lies in the urgency to develop treatments rapidly to address the pressing global health crisis. Traditional drug development processes typically require extensive preclinical testing, followed by multiple phases of clinical trials to establish safety and efficacy. The need for accelerated timelines in pandemic situations necessitates innovative strategies to streamline these processes without compromising scientific rigor. Identifying suitable drug candidates for repurposing presents its own set of challenges.120–122 Repurposing involves investigating existing drugs approved for other conditions to determine their effectiveness against the new target, in this case, SARS-CoV-2. While this approach leverages existing knowledge about drug safety profiles, dosages and potential side effects, it requires a deep understanding of the virus’s biology and the specific cellular processes it exploits. The lack of detailed knowledge about the novel virus early in the outbreak complicates the selection of suitable repurposed drugs.
The genetic variability of the virus also poses a challenge in drug development.123 SARS-CoV-2 has shown the ability to mutate and give rise to different variants, some of which may exhibit altered susceptibility to antiviral agents or monoclonal antibodies.124–126 The D614G mutation in the spike protein of SARS-CoV-2 enhances the virus’s ability to spread from person to person, making it more transmissible.127,128 However, this mutation is also associated with a reduction in the virus’s virulence, or its ability to cause severe disease. Through natural selection, variants of SARS-CoV-2 carrying the D614G mutation have become more prevalent due to their increased transmissibility, potentially contributing to the overall spread of the virus in the population. As the virus evolves, the efficacy of initially promising drugs may be compromised. This necessitates continuous monitoring and adaptation of drug development strategies to account for emerging variants and their potential impact on therapeutic interventions. Another challenge is the risk of off-target effects and unintended consequences during drug development and repurposing.128 Drugs designed for one purpose may interact with other cellular components, leading to unforeseen side effects or complications. Rigorous preclinical and clinical testing is essential to identify and mitigate these risks. Additionally, drug interactions with existing medications or underlying health conditions must be carefully considered to ensure the safety and well-being of patients.
The ethical considerations of conducting clinical trials during a pandemic present a complex challenge.129 Balancing the urgency to find effective treatments with the need for rigorous scientific evaluation and informed consent is crucial. Rapid recruitment of participants, especially in severely affected regions, requires careful ethical oversight to ensure that research participants are adequately informed, protected and treated ethically throughout the trial process. Financial and logistical constraints further complicate drug development efforts.129 Allocating resources for large-scale clinical trials, manufacturing and distribution of potential therapeutics demands significant financial investment and global collaboration. Access to experimental treatments and therapeutics must be equitable, addressing issues of affordability and availability on a global scale.
Vaccine research
Vaccine research for COVID-19 has been a global priority since the emergence of the pandemic. Scientists and pharmaceutical companies worldwide have collaborated to develop safe and effective vaccines to combat the spread of the virus. Utilizing various innovative technologies such as mRNA, viral vector, protein subunit and inactivated virus platforms, multiple vaccines have been swiftly developed and authorized for emergency use (Table 2). These vaccines have undergone rigorous clinical trials to assess their safety, efficacy and ability to induce robust immune responses against SARS-CoV-2. The successful development and deployment of COVID-19 vaccines represent a remarkable achievement in the field of vaccinology, offering hope for controlling the pandemic and returning to normalcy worldwide. However, ongoing research efforts continue to address emerging variants of the virus, vaccine distribution challenges and ensuring equitable access to immunization for all populations.
Vaccine Type | Examples | Mechanism of Action | Advantages | Disadvantages |
---|---|---|---|---|
mRNA Vaccines | Pfizer-BioNTech, Moderna | Introduces mRNA encoding spike protein into cells. Cells produce spike protein, triggering immune response. Immune system learns to recognize and attack virus. mRNA degraded after use, no risk of integration. | High efficacy. Rapid development. Non-replicating. No risk of infection. Short-term side effects (e.g., fatigue, headache). | Storage requirements (-70°C for Pfizer-BioNTech). Potential for allergic reactions. |
Viral Vector Vaccines | AstraZeneca, Johnson and Johnson | Uses harmless virus to deliver DNA encoding spike protein. Virus enters cells and delivers genetic material. Cells produce spike protein, triggering immune response. Immune system learns to recognize and attack virus. DNA does not integrate into host genome. | Single-dose option. Easier storage requirements. Induces strong immune response. No risk of infection. Can be stored at standard refrigerator temperatures | Lower efficacy compared to mRNA vaccines. Risk of rare blood clotting events (AstraZeneca). Potential for adenovirus vector immunity. Longer interval between doses (AstraZeneca) |
Protein Subunit Vaccines | Novavax | Introduces harmless spike protein fragments. Immune system learns to recognize and attack virus. | Protein-based, similar to traditional vaccines. Safe and well-tolerated. No risk of infection. Easier storage requirements. | Requires adjuvant for optimal immune response. Lower efficacy compared to mRNA vaccines. Multiple doses may be needed. Delayed availability compared to mRNA vaccines. |
Inactivated Virus Vaccines | Sinovac, Sinopharm | Uses inactivated virus particles. Immune system learns to recognize and attack virus. | Proven technology with a long history of use. Safe and well-tolerated. No risk of infection. Easier storage requirements. | Lower efficacy compared to mRNA vaccines. Requires multiple doses. Potential for vaccine-enhanced disease in some cases. Delayed immune response. |
mRNA vaccine technology and breakthroughs
mRNA vaccine technology represents a groundbreaking approach to vaccination and its success in the development of COVID-19 vaccines has marked a significant milestone in the field of immunology. The Pfizer-BioNTech and Moderna vaccines, both authorized for emergency use against SARS-CoV-2, are pioneering examples of mRNA vaccines.130 The core principle of mRNA vaccines involves introducing a small piece of messenger RNA (mRNA) into the body, encoding a portion of the virus’s genetic information. In the case of the Pfizer-BioNTech and Moderna COVID-19 vaccines, the mRNA encodes the viral spike protein. Once inside human cells, the mRNA instructs the cells to produce the spike protein. The immune system recognizes this foreign protein as a potential threat and mounts a robust immune response, generating both humoral (antibody-mediated) and cellular (T-cell) immunity. Importantly, mRNA vaccines do not contain live or inactivated virus, eliminating the risk of causing the disease they aim to prevent. The Pfizer-BioNTech and Moderna vaccines have demonstrated remarkable immunogenicity, inducing strong and specific immune responses against the SARS-CoV-2 spike protein. Clinical trials have shown high efficacy in preventing symptomatic COVID-19, with both vaccines exceeding significant efficacy. The immune responses elicited by mRNA vaccines have been particularly effective at preventing severe disease, hospitalization and death caused by the virus. Common side effects are generally mild and transient, including pain at the injection site, fatigue, headache and mild flu-like symptoms. Serious adverse events are rare, emphasizing the safety and tolerability of mRNA vaccines.
The durability of protection conferred by mRNA vaccines has been a subject of extensive research. Studies have indicated that the vaccines provide robust protection against COVID-19 for at least six months after completion of the primary vaccination series. However, ongoing research is essential to monitor the long-term durability of protection and assess the need for booster doses. The emergence of new variants of SARS-CoV-2 has also prompted investigations into the effectiveness of mRNA vaccines against these variants, with evidence suggesting maintained protection against severe outcomes. One study conducted meta-analysis to shed light on the effectiveness of vaccination against specific variants of the virus.131 The analysis includes vaccines from different manufacturers, such as Moderna, Pfizer, AstraZeneca, Bharat and Johnson and Johnson, focusing on their performance against different variants of the virus. The meta-analysis found that the Moderna mRNA-based vaccine showed the highest total effectiveness after the first dose compared to other vaccines studied.
While Pfizer showed higher efficacy after both the necessitating updates and booster doses. Coordinating the distribution of updated vaccines and booster shots globally, especially in the face of supply chain challenges, further complicates equitable access. Addressing the dynamics of virus evolution and variant spread requires ongoing surveillance and a flexible approach to vaccine distribution strategies. Ensuring vaccine acceptance and overcoming vaccine hesitancy is crucial for the success of global vaccination campaigns.132 Cultural, social and political factors contribute to hesitancy and effective communication strategies are required to address misinformation and build public trust. Collaborative efforts between governments, healthcare professionals and community leaders are vital in promoting vaccine literacy and encouraging vaccine uptake.
Vector-based vaccines
Vector-based vaccines, utilizing adenovirus vectors, have played a pivotal role in the global effort to combat the COVID-19 pandemic.133,134 AstraZeneca and Johnson and Johnson are prominent examples of vaccines that employ adenovirus vectors to deliver genetic material encoding the spike protein of SARS-CoV-2, triggering an immune response. Adenovirus vectors, in this context, act as vehicles to transport the viral genetic material into human cells, initiating the production of the spike protein and eliciting an immune response.
The AstraZeneca vaccine, developed in collaboration with the University of Oxford, utilizes a Chimpanzee Adenovirus vector (ChAdOx1). Similarly, the Johnson and Johnson vaccine employs a human Adenovirus type 26 (Ad26) vector.135 These vaccines have demonstrated efficacy in preventing COVID-19 and, more importantly, in reducing severe disease, hospitalization and mortality. However, the side effects associated with the AstraZeneca vaccine, as a case study reported.136 is a big concern which needs further research in vaccine development. Common side effects were reported after receiving both the first and second doses of the vaccine. Pain at the injection site was the most frequently reported side effect, followed by swelling and fever after the first dose. After the second dose, adverse effects included headache, swelling and burning at the injection site. One advantage of vector-based vaccines is their ability to generate robust and broad immune responses, involving both antibody-mediated and cellular immunity.137 This comprehensive immune response is crucial in providing protection against different variants of SARS-CoV-2. Studies have indicated that these vaccines maintain efficacy against several variants, including the Alpha, Beta and Delta variants, although there may be some reduction in effectiveness against certain variants.138,139 Ongoing research and surveillance are essential to monitor the impact of emerging variants on vaccine efficacy and inform potential adjustments to vaccination strategies.
Booster strategies for vector-based vaccines have become a focus of research and public health efforts, especially in the context of waning immunity and the emergence of new variants. Studies and real-world data suggest that booster doses significantly enhance and prolong the protective immune response,133 Booster shots are designed to reinforce and expand the immune memory, providing an additional layer of defense against SARS-CoV-2. The choice of booster vaccine may vary, with some individuals receiving a different vaccine than their initial series, known as mix-and-match or heterologous boosting.140 This approach aims to optimize the immune response by leveraging different vaccine platforms. Logistical considerations, such as storage requirements and distribution challenges, also influence the deployment of vector-based vaccines.141 Unlike mRNA vaccines, which require ultra-cold storage, vector-based vaccines are often more stable at standard refrigeration temperatures. This characteristic simplifies distribution and facilitates vaccine accessibility, particularly in regions with limited access to specialized storage facilities.
Despite the successes of vector-based vaccines, challenges and concerns have arisen, including rare instances of blood clotting disorders associated with the AstraZeneca vaccine.142,143 Regulatory agencies and healthcare professionals closely monitor and evaluate these potential side effects to ensure the overall safety of vaccination programs. Addressing public concerns through transparent communication and providing clear risk-benefit assessments are crucial elements in maintaining public confidence in vaccination efforts. Ongoing research into booster strategies, monitoring vaccine efficacy against emerging variants and addressing safety concerns contribute to optimizing the use of these vaccines in diverse populations. The adaptability, stability and immunogenicity of vector-based vaccines contribute to their significant role in expanding global vaccination coverage and mitigating the impact of the pandemic.
Challenges in vaccine distribution and global equitable access
The global distribution of COVID-19 vaccines faces a myriad of challenges, hindering efforts to ensure equitable access on a global scale. One major obstacle is the stark imbalance in vaccine distribution between high-income and low-income countries. Wealthier nations secured a significant portion of available vaccine doses through pre-purchase agreements with pharmaceutical companies, leaving less affluent nations with limited access.144 This imbalance exacerbates global health disparities and poses a substantial hurdle in achieving widespread immunity.
Logistical challenges further complicate vaccine distribution efforts. The storage and transportation requirements of certain vaccines, such as those needing ultra-cold storage like the Pfizer-BioNTech vaccine, pose difficulties, especially in regions lacking the necessary infrastructure. Maintaining the cold chain during distribution to remote or underserved areas becomes a logistical challenge that demands coordinated efforts from governments, international organizations and pharmaceutical companies to ensure the vaccines reach their destinations in optimal condition.145 Global vaccine production capacity is another bottleneck in achieving equitable access.146 The complex manufacturing processes, limited production facilities and a shortage of raw materials contribute to a slow pace of vaccine production. Expanding manufacturing capabilities and establishing partnerships between pharmaceutical companies and manufacturers in different regions are essential steps to increase the overall vaccine supply and facilitate more equitable distribution.
The emergence of new variants of the virus adds another layer of complexity to vaccine distribution efforts. Some variants may impact the effectiveness of existing vaccines, necessitating updates or booster doses. Coordinating the distribution of updated vaccines and booster shots globally, especially in the face of supply chain challenges, further complicates equitable access.147 Addressing the dynamics of virus evolution and variant spread requires ongoing surveillance and a flexible approach to vaccine distribution strategies. Ensuring vaccine acceptance and overcoming vaccine hesitancy is crucial for the success of global vaccination campaigns.132 Cultural, social and political factors contribute to hesitancy and effective communication strategies are required to address misinformation and build public trust. Collaborative efforts between governments, healthcare professionals and community leaders are vital in promoting vaccine literacy and encouraging vaccine uptake. The Access to COVID-19 Tools Accelerator (ACT-A) and its vaccine arm, COVAX, represent global initiatives aimed at promoting equitable access to vaccines.26 COVAX, co-led by Gavi, the Coalition for Epidemic Preparedness Innovations (CEPI) and the World Health Organization (WHO), seeks to distribute vaccines to low- and middle-income countries.148 However, challenges persist, including funding gaps, supply chain disruptions and the need for increased collaboration between governments, pharmaceutical companies and international organizations. Addressing these challenges requires a comprehensive, coordinated and multilateral approach. The international community must prioritize the removal of barriers to vaccine access, enhance manufacturing capacities, improve distribution infrastructure and engage in transparent and equitable allocation mechanisms. Collaborative efforts to support technology transfer and knowledge sharing can further contribute to building sustainable vaccine production capabilities in different regions.
Future directions
Future research directions in the battle against COVID-19 encompass a spectrum of unanswered questions, knowledge gaps and areas requiring further investigation. One critical aspect involves the long-term effects of SARS-CoV-2 infection, commonly referred to as “long COVID” or post-acute sequelae of SARS-CoV-2 infection. Understanding the mechanisms underlying persistent symptoms, organ involvement and the impact on patients’ quality of life remains a priority. Unraveling the complexities of long-term effects requires longitudinal studies, molecular investigations and comprehensive clinical assessments to guide targeted interventions and support strategies. Recent studies highlight the scarcity of experimental studies investigating interactions between SARS-CoV-2 and other pathogens, particularly in animal models.149,150 However, existing research indicates that coinfections with SARS-CoV-2 and influenza can exacerbate the severity of COVID-19. The effects of coinfection on viral loads varied across studies, possibly due to differing methodologies. Although previous influenza vaccination appears to reduce the risk of COVID-19, its role in interaction remains uncertain. Limited research has explored the impact of other pathogens such as Respiratory Syncytial Virus (RSV) and rhinoviruses, underscoring the need for further investigation into interactions with endemic bacteria. Overall, there is an urgent need for additional experimental and epidemiological studies to better understand SARS-CoV-2 interactions with other pathogens.
The potential for cross-disciplinary collaborations emerges as a key avenue for advancing our understanding of COVID-19. Integrating expertise from virology, immunology, epidemiology and other disciplines can catalyze innovative approaches to address complex challenges. For instance, collaborations between basic scientists and clinicians can bridge gaps between laboratory findings and clinical applications, facilitating the translation of research discoveries into effective therapeutic strategies. Additionally, collaborations with data scientists and computational biologists can enhance the analysis of large datasets, uncovering patterns and associations that inform both treatment and prevention efforts. Advancements in diagnostics, treatments and vaccines continue to be pivotal in shaping the trajectory of the pandemic. Novel technologies and emerging approaches, such as next-generation sequencing for variant detection, CRISPR-based diagnostics and artificial intelligence applications in vaccine design, represent promising frontiers. These technologies can enhance the speed, accuracy and specificity of diagnostics, allowing for real-time monitoring and intervention. Similarly, advancements in treatments, including antiviral agents and immunomodulatory therapies, demand ongoing research to optimize their use in different patient populations and disease stages.
Integrating research findings into public health strategies is critical for effective pandemic management. This involves not only disseminating scientific knowledge but also implementing evidence-based interventions at the population level. Strategies for vaccination campaigns, therapeutics distribution and public health messaging should be informed by the latest research on vaccine effectiveness, treatment outcomes and epidemiological trends. Collaborative efforts between researchers, public health agencies and policymakers are essential to ensure a seamless translation of scientific advancements into practical and impactful public health measures.
CONCLUSION
The journey through the comprehensive scientific review of COVID-19 has highlighted several key research developments that have significantly advanced our understanding of the virus and shaped our strategies for prevention, treatment and mitigation. Noteworthy developments include the unprecedented speed in vaccine development, exemplified by the success of mRNA and vector-based vaccines from Pfizer-BioNTech, Moderna, AstraZeneca and Johnson and Johnson. The evolution of SARS-CoV-2, characterized by the emergence of variants of concern, has underscored the importance of ongoing genomic surveillance and adaptive vaccine strategies. Advances in diagnostics, therapeutics and immunomodulatory treatments have played crucial roles in managing the impact of the virus on public health.
Research efforts have elucidated key aspects of SARS-CoV-2 biology, including the spike protein mutations, receptor binding domain alterations and various immune evasion strategies employed by the virus. Insights into host-virus interactions and immune response dynamics have provided a foundation for understanding disease severity, asymptomatic transmission and genetic factors influencing susceptibility. Investigations into long-term effects and post-acute sequelae have shed light on the multifaceted nature of COVID-19 and its potential implications for individuals beyond the acute phase of infection. The lessons learned from the COVID-19 pandemic have profound implications for future pandemic preparedness and response. The rapid development and deployment of vaccines showcase the power of collaborative efforts, technological innovation and adaptive regulatory processes. Strengthening global surveillance systems, enhancing diagnostic capabilities and investing in research infrastructure are imperative for early detection and containment of emerging infectious diseases. The significance of equitable vaccine distribution has underscored the need for international collaboration, resource-sharing and addressing health disparities to ensure that all nations have access to life-saving interventions.
The dynamic nature of the virus and the continuous emergence of variants emphasize the necessity of flexible and adaptable strategies. Future pandemic preparedness should focus on developing platform technologies for vaccine design, therapeutics and diagnostics that can be rapidly deployed against novel pathogens. International cooperation in sharing data, research findings and resources is crucial for a coordinated global response. The integration of diverse expertise, including virology, immunology, epidemiology and data science, is essential for a comprehensive understanding of emerging infectious diseases. As we conclude this review, a resounding call to action echoes through the scientific community. Continued research and collaboration are imperative to address remaining uncertainties, fill knowledge gaps and refine strategies for managing COVID-19 and future pandemics. Ongoing surveillance of SARS-CoV-2 variants, comprehensive investigations into the long-term effects of the virus and efforts to optimize treatment protocols are essential components of this collective endeavor. International collaboration and knowledge-sharing platforms must be strengthened to facilitate the rapid exchange of information, research findings and resources. Investments in research infrastructure, including genomics, diagnostics and clinical trials, are necessary to build resilience against emerging infectious threats. The scientific community, policymakers and funding agencies must sustain their commitment to supporting research initiatives that contribute to the ongoing fight against COVID-19 and bolster global preparedness for future pandemics.
In conclusion, the scientific landscape surrounding COVID-19 has witnessed remarkable progress, but challenges persist. The journey ahead demands unwavering dedication, collaboration and a commitment to advancing knowledge for the benefit of global health. As we navigate the complexities of infectious diseases, the lessons learned from COVID-19 serve as a compass guiding us toward a future where research and collaboration stand as formidable pillars in our defense against pandemics.
Cite this article:
Alzahrani A. Research Developments and Understandings in the Dynamics of COVID-19: A Comprehensive Review. Int. J. Pharm. Investigation. 2024;14(4):1096-114.
ACKNOWLEDGEMENT
The author thanks to Department of Applied Medical Sciences, Applied College, Al-Baha University, Al-Baha City and Kingdom of Saudi Arabia.
ABBREVIATIONS
SARS-CoV-2 | Severe Acute Respiratory Syndrome Coronavirus 2 |
---|---|
RBD | Receptor binding domain |
ACE2 | Angiotensin-Converting Enzyme 2 |
S | Spike |
E | Envelope |
M | Membrane |
N | Nucleocapsid |
ssRNA | Single-stranded RNA |
DMVs | Double-membrane Vesicles |
CMs | Convoluted Membranes |
DMSs | Double-membrane Spherules |
sg mRNAs | Subgenomic mRNAs |
ER | Endoplasmic Reticulum |
ERGIC | ER-to-Golgi Intermediate Compartment |
Ad26 | Adenovirus type 26 |
scRNA-seq | Single-cell RNA sequencing |
CEPI | Coalition for Epidemic Preparedness Innovations |
References
- Krishnamoorthy S, Swain B, Verma RS, Gunthe SS. SARS-CoV, MERS-CoV and 2019-nCoV viruses: an overview of origin, evolution and genetic variations. Virusdisease. 2020;31(4):411-23. [PubMed] | [CrossRef] | [Google Scholar]
- Hatmal MM, Alshaer W, Al-Hatamleh MA, Hatmal M, Smadi O, Taha MO, et al. Comprehensive structural and molecular comparison of spike proteins of SARS-CoV-2, SARS-CoV and MERS-CoV and their interactions with ACE2. Cells. 2020;9(12):2638 [PubMed] | [CrossRef] | [Google Scholar]
- Rabaan AA, Mutair AA, Alawi ZA, Alhumaid S, Mohaini MA, Aldali J, et al. Comparative pathology, molecular pathogenicity, immunological features and genetic characterization of three highly pathogenic human coronaviruses (MERS-CoV, SARS-CoV and SARS-CoV-2). European Review for Medical and Pharmacological Sciences. 2021;25(22):7162-84. [PubMed] | [CrossRef] | [Google Scholar]
- Hedman HD, Krawczyk E, Helmy YA, Zhang L, Varga CJP. Host diversity and potential transmission pathways of SARS-CoV-2 at the human-animal interface. Pathogens. 2021;10(2):180 [PubMed] | [CrossRef] | [Google Scholar]
- Yadav T, Saxena SK. Pathogenesis, Diagnosis, and Therapeutics, Transmission cycle of SARS-CoV and SARS-CoV-2. Coronavirus Disease 2019 (COVID-19). 2020:33-42. [CrossRef] | [Google Scholar]
- Mohammad T, Choudhury A, Habib I, Asrani P, Mathur Y, Umair M, et al. Genomic variations in the structural proteins of SARS-CoV-2 and their deleterious impact on pathogenesis: a comparative genomics approach. Frontiers in cellular and infection microbiology. 2021;11:765039 [PubMed] | [CrossRef] | [Google Scholar]
- Gupta D, Sharma P, Singh M, Kumar M, Ethayathulla A, Kaur P, et al. Structural and functional insights into the spike protein mutations of emerging SARS-CoV-2 variants. Cellular and Molecular Life Sciences. 2021(24):7967-89. [PubMed] | [CrossRef] | [Google Scholar]
- Mehra R. and Kepp KP. Structure and mutations of SARS-CoV-2 spike protein: a focused overview. ACS Infectious Diseases. 2021;8(1):29-58. Epub 2021 Dec 2 [PubMed] | [CrossRef] | [Google Scholar]
- Romano M, Ruggiero A, Squeglia F, Maga G, Berisio R. A structural view of SARS-CoV-2 RNA replication machinery: RNA synthesis, proofreading and final capping. Cells. 2020;9(5):1267 [PubMed] | [CrossRef] | [Google Scholar]
- Primorac D, Vrdoljak K, Brlek P, Pavelić E, Molnar V, Matišić V, et al. Adaptive immune responses and immunity to SARS-CoV-2. Frontiers in Immunology. 2022;13:848582 [PubMed] | [CrossRef] | [Google Scholar]
- Cohen KW, Linderman SL, Moodie Z, Czartoski J, Lai L, Mantus G, et al. Longitudinal analysis shows durable and broad immune memory after SARS-CoV-2 infection with persisting antibody responses and memory B and T cells. Cell Reports Medicine. 2021;2(7):100354 [PubMed] | [CrossRef] | [Google Scholar]
- Otto SP, Day T, Arino J, Colijn C, Dushoff J, Li M, et al. The origins and potential future of SARS-CoV-2 variants of concern in the evolving COVID-19 pandemic. Current Biology. 2021;31(14) [PubMed] | [CrossRef] | [Google Scholar]
- Sanyaolu A, Okorie C, Marinkovic A, Haider N, Abbasi AF, Jaferi U, et al. The emerging SARS-CoV-2 variants of concern. Therapeutic Advances in Infectious Disease. 2021;8:20499361211024372 [PubMed] | [Google Scholar]
- Parlikar A, Kalia K, Sinha S, Patnaik S, Sharma N, Vemuri SG, Sharma G, et al. Understanding genomic diversity, pan-genome and evolution of SARS-CoV-2. PeerJ. 2020;8 eCollection 2020 [PubMed] | [CrossRef] | [Google Scholar]
- Kissler SM, Tedijanto C, Goldstein E, Grad YH, Lipsitch M. Projecting the transmission dynamics of SARS-CoV-2 through the postpandemic period. Science. 2020;368(6493):860-8. [PubMed] | [CrossRef] | [Google Scholar]
- Tsai P-H, Lai W-Y, Lin Y-Y, Luo Y-H, Lin Y-T, Chen H-K, et al. Clinical manifestation and disease progression in COVID-19 infection. Journal of the Chinese Medical Association. 2021;84(1):3-8. [PubMed] | [CrossRef] | [Google Scholar]
- Ghasemi S, Harmooshi NN, Rahim F. Diagnostic utility of antigen detection rapid diagnostic tests for Covid-19: a systematic review and meta-analysis. Diagnostic Pathology. 2022;17(1):36 [PubMed] | [Google Scholar]
- Beyer DK, Forero A. Mechanisms of antiviral immune evasion of SARS-CoV-2. Journal of Molecular Biology. 2022;434(6):167265 [PubMed] | [CrossRef] | [Google Scholar]
- Minkoff JM, tenOever B. Innate immune evasion strategies of SARS-CoV-2. Nature Reviews Microbiology. 2023;21(3):178-94. [PubMed] | [CrossRef] | [Google Scholar]
- Papi M, Pozzi D, Palmieri V, Caracciolo G. Principles for optimization and validation of mRNA lipid nanoparticle vaccines against COVID-19 using 3D bioprinting. Nano Today. 2022;43:101403 [PubMed] | [CrossRef] | [Google Scholar]
- Lotfi H, Mazar MG, Ei NM, Fahim M, Yazdi NS. Vaccination is the most effective and best way to avoid the disease of COVID-19. Immunity, Inflammation and Disease. 2023;11(8) [PubMed] | [CrossRef] | [Google Scholar]
- Delardas O, Kechagias KS, Pontikos PN, Giannos P. Socio-Economic impacts and challenges of the coronavirus pandemic (COVID-19): an updated review. Sustainability. 2022;14(15):9699 [CrossRef] | [Google Scholar]
- Pal M, Kerorsa GB, Kandi V. A knowledge update on SARS-Coronavirus-2 (SARS-CoV-2)/COVID-19 and its global public health implications. American Journal of Clinical Medicine Research. 2022;8(1):23-27. [CrossRef] | [Google Scholar]
- Chiu KH-Y, Sridhar S, Yuen K-Y. Preparation for the next pandemic: challenges in strengthening surveillance. Emerging Microbes and Infections. 2023;12(2):2240441 [PubMed] | [CrossRef] | [Google Scholar]
- Siegel RM, Mallow PJ. The impact of COVID-19 on vulnerable populations and implications for children and health care policy. Clinical Pediatrics. 2021;60(2):93-8. [PubMed] | [CrossRef] | [Google Scholar]
- Eccleston-Turner M, Upton H. International collaboration to ensure equitable access to vaccines for COVID-19: the ACT-Accelerator and the COVAX facility. The Milbank Quarterly. 2021;99(2):426-49. [PubMed] | [CrossRef] | [Google Scholar]
- Both LM, Zoratto G, Calegaro VC, Ramos-Lima LF, Negretto BL, Hauck S, et al. COVID-19 pandemic and social distancing: economic, psychological, family and technological effects. Trends in Psychiatry and Psychotherapy. 2021;43(2):85-91. [PubMed] | [CrossRef] | [Google Scholar]
- Dykstra MP, Baitchman EJ. A call for one health in medical education: How the COVID-19 pandemic underscores the need to integrate human, animal and environmental health. Academic Medicine. 2021;96(7):951-3. [PubMed] | [CrossRef] | [Google Scholar]
- Flores-Vega VR, Monroy-Molina JV, Jiménez-Hernández LE, Torres AG, Santos-Preciado JI, Rosales-Reyes R, et al. SARS-CoV-2: Evolution and emergence of new viral variants. Viruses. 2022;14(4):653 [PubMed] | [CrossRef] | [Google Scholar]
- Muñoz-Fontela C, Widerspick L, Albrecht RA, Beer M, Carroll MW, de Wit E, et al. Advances and gaps in SARS-CoV-2 infection models. PLOS Pathogens. 2022;18(1) [PubMed] | [CrossRef] | [Google Scholar]
- Borkotoky S, Dey D, Hazarika Z. Interactions of angiotensin-converting enzyme-2 (ACE2) and SARS-CoV-2 spike receptor-binding domain (RBD): A structural perspective. Molecular Biology Reports. 2023;50(3):2713-21. [PubMed] | [CrossRef] | [Google Scholar]
- Naresh GK, Guruprasad L. Mutations in the receptor-binding domain of human SARS CoV-2 spike protein increases its affinity to bind human ACE-2 receptor. Journal of Biomolecular Structure and Dynamics. 2023;41(6):2368-81. [PubMed] | [CrossRef] | [Google Scholar]
- Banerjee S, Wang X, Du S, Zhu C, Jia Y, Wang Y, Cai Q, et al. Comprehensive role of SARS-CoV-2 spike glycoprotein in regulating host signaling pathway. Journal of Medical Virology. 2022;94(9):4071-87. [PubMed] | [CrossRef] | [Google Scholar]
- Chakraborty S. Evolutionary and structural analysis elucidates mutations on SARS-CoV2 spike protein with altered human ACE2 binding affinity. Biochemical and Biophysical Research Communications. 2021;538:97-103. [PubMed] | [CrossRef] | [Google Scholar]
- Lupala CS, Ye Y, Chen H, Su X-D, Liu H. Mutations on RBD of SARS-CoV-2 Omicron variant result in stronger binding to human ACE2 receptor. Biochemical and Biophysical Research Communications. 2022;590:34-41. [PubMed] | [CrossRef] | [Google Scholar]
- Duong D. Alpha, Beta, Delta, Gamma: What’s important to know about SARS-CoV-2 variants of concern?. Canadian Medical Association Journal. 2021;193(27) [PubMed] | [CrossRef] | [Google Scholar]
- Liu H, Wei P, Zhang Q, Aviszus K, Linderberger J, Yang J, et al. The Lambda variant of SARS-CoV-2 has a better chance than the Delta variant to escape vaccines. bioRxiv. 2021:457692 [PubMed] | [CrossRef] | [Google Scholar]
- Rees-Spear C, Muir L, Griffith SA, Heaney J, Aldon Y, Snitselaar JL, et al. The effect of spike mutations on SARS-CoV-2 neutralization. Cell Reports. 2021;34(12):108890 [PubMed] | [CrossRef] | [Google Scholar]
- Chen X, Chen Z, Azman AS, Sun R, Lu W, Zheng N, et al. Neutralizing antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants induced by natural infection or vaccination: a systematic review and pooled analysis. Clinical Infectious Diseases. 2022;74(4):734-42. [PubMed] | [CrossRef] | [Google Scholar]
- Ma W, Fu H, Jian F, Cao Y, Li M. Immune evasion and ACE2 binding affinity contribute to SARS-CoV-2 evolution. Nature Ecology and Evolution. 2023;7(9):1457-66. [PubMed] | [CrossRef] | [Google Scholar]
- Vkovski P, Kratzel A, Steiner S, Stalder H, Thiel V. Coronavirus biology and replication: implications for SARS-CoV-2. Nature Reviews Microbiology. 2021;19(3):155-70. [PubMed] | [CrossRef] | [Google Scholar]
- Lazarevic I, Pravica V, Miljanovic D, Cupic M. Immune evasion of SARS-CoV-2 emerging variants: what have we learnt so far?. Viruses. 2021;13(7):1192 [PubMed] | [CrossRef] | [Google Scholar]
- Mantlo E, Bukreyeva N, Maruyama J, Paessler S, Huang C. Antiviral activities of type I interferons to SARS-CoV-2 infection. Antiviral Research. 2020;179:104811 [PubMed] | [CrossRef] | [Google Scholar]
- Forni D, Cagliani R, Pontremoli C, Mozzi A, Pozzoli U, Clerici M, Sironi M, et al. Antigenic variation of SARS-CoV-2 in response to immune pressure. Molecular Ecology. 2021;30(14):3548-59. [PubMed] | [CrossRef] | [Google Scholar]
- Cariou M, Picard L, Guéguen L, Jacquet S, Cimarelli A, Fregoso OI, et al. Distinct evolutionary trajectories of SARS-CoV-2-interacting proteins in bats and primates identify important host determinants of COVID-19. Proceedings of the National Academy of Sciences. 2022;119(35) [PubMed] | [CrossRef] | [Google Scholar]
- Gavriatopoulou M, Korompoki E, Fotiou D, Ntanasis-Stathopoulos I, Psaltopoulou T, Kastritis E, et al. Organ-specific manifestations of COVID-19 infection. Clinical and Experimental Medicine. 2020;20(4):493-506. [PubMed] | [CrossRef] | [Google Scholar]
- Perez-Gomez R. The development of SARS-CoV-2 variants: the gene makes the disease. Journal of Developmental Biology. 2021;9(4):58 [PubMed] | [CrossRef] | [Google Scholar]
- Bushman M, Kahn R, Taylor BP, Lipsitch M, Hanage WP. Population impact of SARS-CoV-2 variants with enhanced transmissibility and/or partial immune escape. Cells. 2021;184(26):6229-42. [PubMed] | [CrossRef] | [Google Scholar]
- Tyagi N, Sardar R, Gupta D. Natural selection plays a significant role in governing the codon usage bias in the novel SARS-CoV-2 variants of concern (VOC). PeerJ. 2022;10 [PubMed] | [CrossRef] | [Google Scholar]
- Jarvis MC. Aerosol transmission of SARS-CoV-2: physical principles and implications. Frontiers in Public Health. 2020;8:590041 [PubMed] | [CrossRef] | [Google Scholar]
- Zhou M, Zou J. A dynamical overview of droplets in the transmission of respiratory infectious diseases. Physics of Fluids. 2021;33(3):031301 [PubMed] | [CrossRef] | [Google Scholar]
- Pöhlker ML, Pöhlker C, Krüger OO, Förster J-D, Berkemeier T, Elbert W, et al. Respiratory aerosols and droplets in the transmission of infectious diseases. American Physical Society. 2023;95(4):045001 [CrossRef] | [Google Scholar]
- Kumar S, Singh R, Kumari N, Karmakar S, Behera M, Siddiqui AJ, et al. Current understanding of the influence of environmental factors on SARS-CoV-2 transmission, persistence and infectivity. Environmental Science and Pollution Research. 2021;28:6267-88. [PubMed] | [CrossRef] | [Google Scholar]
- Mao N, Zhang D, Li Y, Li Y, Li J, Zhao L, et al. How do temperature, humidity and air saturation state affect the COVID-19 transmission risk?. Environmental Science and Pollution Research. 2023;30(2):3644-58. [PubMed] | [CrossRef] | [Google Scholar]
- Busato F, Cavallini A. Window Opening or Mechanical Ventilation Systems for Italian Schools? Energetic Aspects, CO2 Concentration and Infection Risk Assessment Based on the SARS-CoV-2 Data. buildings. 2023;13(7):1743 [CrossRef] | [Google Scholar]
- Song B-M, Lee G-H, Han H-J, Yang J-H, Lee E-G, Gu H, et al. Ultraviolet-C light at 222 nm has a high disinfecting spectrum in environments contaminated by infectious pathogens, including SARS-CoV-2. PLoS One. 2023;18(11) [PubMed] | [CrossRef] | [Google Scholar]
- Boterman WJUS. Population density and SARS-CoV-2 pandemic: Comparing the geography of different waves in the Netherlands. Urban studies. 2023;60(8):1377-402. [PubMed] | [CrossRef] | [Google Scholar]
- Zhang G, Poslad S, Fan Y, Rui X. Quantitative spatiotemporal impact of dynamic population density changes on the COVID-19 pandemic in China’s mainland. Geo-spatial Information Science. 2023;26(4):642-63. [CrossRef] | [Google Scholar]
- Yang J, Sun D, Xia T, Shi S, Suo J, Kuang H, et al. Monitoring Prevalence and Persistence of Environmental Contamination by SARS-CoV-2 RNA in a Makeshift Hospital for Asymptomatic and Very Mild COVID-19 Patients. International Journal of Public Health. 2023;68:1605994 [PubMed] | [CrossRef] | [Google Scholar]
- Wang L, Didelot X, Yang J, Wong G, Shi Y, Liu W, et al. Inference of person-to-person transmission of COVID-19 reveals hidden super-spreading events during the early outbreak phase. Nature Communications. 2020;11(1):5006 [PubMed] | [CrossRef] | [Google Scholar]
- Goyal A, Reeves DB, Cardozo-Ojeda EF, Schiffer JT, Mayer B. Viral load and contact heterogeneity predict SARS-CoV-2 transmission and super-spreading events. eLife. 2021;10 [PubMed] | [CrossRef] | [Google Scholar]
- Müller J, Hösel V. Contact tracing and super-spreaders in the branching-process model. Journal of Mathematical Biology. 2023;86(2):24 [PubMed] | [CrossRef] | [Google Scholar]
- Qiu X, Nergiz AI, Maraolo AE, Bogoch II, Low N, Cevik M, et al. The role of asymptomatic and pre-symptomatic infection in SARS-CoV-2 transmission-a living systematic review. Clinical Microbiology and Infection. 2021;27(4):511-9. [PubMed] | [CrossRef] | [Google Scholar]
- Sah P, Fitzpatrick MC, Zimmer CF, Abdollahi E, Juden-Kelly L, Moghadas SM, et al. Asymptomatic SARS-CoV-2 infection: A systematic review and meta-analysis. Proceedings of the National Academy of Sciences. 2021;118(34) [PubMed] | [Google Scholar]
- Almadhi MA, Abdulrahman A, Sharaf SA, AlSaad D, Stevenson NJ, Atkin SL, AlQahtani MM, et al. The high prevalence of asymptomatic SARS-CoV-2 infection reveals the silent spread of COVID-19. International Journal of Infectious Diseases. 2021;105:656-61. [PubMed] | [CrossRef] | [Google Scholar]
- Casey M, Griffin J, McAloon CG, Byrne AW, Madden JM, Mc Evoy D, et al. Estimating pre-symptomatic transmission of COVID-19: a secondary analysis using published data. medRxiv. 2020:20094870 [CrossRef] | [Google Scholar]
- Liu Y, Funk S, Flasche S. The contribution of pre-symptomatic infection to the transmission dynamics of COVID-2019. Wellcome Open Research. 2020;5:58 [PubMed] | [CrossRef] | [Google Scholar]
- Mugglestone MA, Ratnaraja NV, Bak A, Islam J, Wilson JA, Bostock J, et al. Presymptomatic, asymptomatic and post-symptomatic transmission of SARS-CoV-2: Joint British infection association (BIA), healthcare infection society (HIS), infection prevention society (IPS) and royal College of pathologists (RCPath) guidance. BMC Infectious Diseases. 2022;22(1):453 [PubMed] | [CrossRef] | [Google Scholar]
- Ebrahimi M, Harmooshi NN, Rahim F. Diagnostic utility of antigen detection rapid diagnostic tests for COVID-19: a systematic review and meta-analysis. BMC Infectious Diseases. 2021;17(1):36 [PubMed] | [CrossRef] | [Google Scholar]
- Núñez I, Belaunzarán-Zamudio PF, Caro-Vega Y. Result turnaround time of RT-PCR for SARS-cov-2 is the main cause of COVID-19 diagnostic delay: A country-wide observational study of mexico and colombia. Revista de Investigación Clínica. 2022;74(2):71-80. [PubMed] | [CrossRef] | [Google Scholar]
- Arevalo-Rodriguez I, Buitrago-Garcia D, Simancas-Racines D, Zambrano-Achig P, Del Campo R, Ciapponi A, et al. False-negative results of initial RT-PCR assays for COVID-19: a systematic review. PLoS One. 2020;5(12) [PubMed] | [CrossRef] | [Google Scholar]
- Mahmood TB, Saha A, Hossan MI, Mizan S, Arman SAS, Chowdhury AS, et al. A next generation sequencing (NGS) analysis to reveal genomic and proteomic mutation landscapes of SARS-CoV-2 in South Asia. Current Research in Microbial Sciences. 2021;2:100065 [PubMed] | [CrossRef] | [Google Scholar]
- Jiang W, Ji W, Zhang Y, Xie Y, Chen S, Jin Y, Duan GJV, et al. An update on detection technologies for SARS-CoV-2 variants of concern. Viruses. 2022;14(11):2324 [PubMed] | [CrossRef] | [Google Scholar]
- Singh RR. Next-generation sequencing in high-sensitive detection of mutations in tumors: challenges, advances and applications. The Journal of Molecular Diagnostics. 2020;22(8):994-1007. [PubMed] | [CrossRef] | [Google Scholar]
- Rabiee N, Bagherzadeh M, Ghasemi A, Zare H, Ahmadi S, Fatahi Y, et al. Point-of-use rapid detection of SARS-CoV-2: nanotechnology-enabled solutions for the COVID-19 pandemic. International Journal of Molecular Sciences. 2020;21(14):5126 [PubMed] | [CrossRef] | [Google Scholar]
- Aguilar-Shea AL, Vera-García M, Güerri-Fernández R. Rapid antigen tests for the detection of SARS-CoV-2: a narrative review. Atención Primaria. 2021;53(9):102127 [PubMed] | [CrossRef] | [Google Scholar]
- Dinnes J, Sharma P, Berhane S, van Wyk SS, Nyaaba N, Domen J, et al. Rapid, point-of-care antigen tests for diagnosis of SARS-CoV-2 infection. Cochrane Database of Systematic Reviews. 2022;7(7) [PubMed] | [CrossRef] | [Google Scholar]
- Iliescu FS, Ionescu AM, Gogianu L, Simion M, Dediu V, Chifiriuc MC, et al. Point-of-Care Testing-The Key in the Battle against SARS-CoV-2 Pandemic. 2021; Micromachines. 2021;12(12):1464 [PubMed] | [CrossRef] | [Google Scholar]
- Nanayakkara L, Pettigrew TR, Overton J, Ryan PL, Pawar AK, Midson HM, et al. Reduction in cycle time for a rapid polymerase chain reaction diagnostic test at the point of care. Journal of Infection Prevention. 2023;24(1):23-9. [PubMed] | [CrossRef] | [Google Scholar]
- Biswas GC, Choudhury S, Rabbani MM, Das J. A review on potential electrochemical point-of-care tests targeting pandemic infectious disease detection: COVID-19 as a reference. Chemosensors. 2022;10(7):269 [CrossRef] | [Google Scholar]
- Zhang S, Ma P, Orzechowski M, Lemmer A, Rzasa K, Bagnall J, et al. High-throughput neutralization and serology assays reveal correlated but highly variable humoral immune responses in a large population of individuals infected with SARS-CoV-2 in the US between March and August 2020. mBio. 2023;14(2) [PubMed] | [CrossRef] | [Google Scholar]
- Garcia-Fogeda I, Besbassi H, Larivière Y, Ogunjimi B, Abrams S, Hens N, et al. Within-host modeling to measure dynamics of antibody responses after natural infection or vaccination: A systematic review. Vaccines. 2023;41(25):3701-9. [PubMed] | [CrossRef] | [Google Scholar]
- Swartz MD, DeSantis SM, Yaseen A, Brito FA, Valerio-Shewmaker MA, Messiah SE, et al. Antibody duration after infection from SARS-CoV-2 in the Texas coronavirus antibody response survey. The Journal of Infectious Diseases. 2023;227(2):193-201. [PubMed] | [CrossRef] | [Google Scholar]
- Wang M, Zhou B, Fan Q, Zhou X, Liao X, Lin J, et al. Infections, Omicron variants escape the persistent SARS-CoV-2-specific antibody response in 2-year COVID-19 convalescents regardless of vaccination. Emerging Microbes and Infections. 2023;12(1):2151381 [PubMed] | [CrossRef] | [Google Scholar]
- Painter MM, Johnston TS, Lundgreen KA, Santos JJ, Qin JS, Goel RR, et al. Williams J.C.J.N.I., Prior vaccination promotes early activation of memory T cells and enhances immune responses during SARS-CoV-2 breakthrough infection. Nature Immunology. 2023;24(10):1711-24. [PubMed] | [CrossRef] | [Google Scholar]
- Le Bert N, Samandari T. Silent battles: immune responses in asymptomatic SARS-CoV-2 infection. Cellular and Molecular Immunology. 2024(2):159-170. [PubMed] | [CrossRef] | [Google Scholar]
- Gonzalez-Kozlova E. Molecular profiling of liquid biopsies for precision oncology. Advances in Experimental Medicine and Biology. 2022:235-47. [PubMed] | [CrossRef] | [Google Scholar]
- Franco-Luiz APM, Fernandes NMGS, de Souza Silva TB, Bernardes WPdOS, Westin MR, Santos TG, et al. Longitudinal study of humoral immunity against SARS-CoV-2 of health professionals in Brazil: the impact of booster dose and reinfection on antibody dynamics. Frontiers in Immunology. 2023;14:1220600 [PubMed] | [CrossRef] | [Google Scholar]
- Patalon T, Saciuk Y, Perez G, Peretz A, Ben-Tov A, Gazit S, et al. Dynamics of naturally acquired immunity against severe acute respiratory syndrome coronavirus 2 in children and adolescents. The Journal of Pediatrics. 2023:113371 [PubMed] | [CrossRef] | [Google Scholar]
- de León P, Cañas-Arranz R, Bustos MJ, Sáiz M, Sobrino F. Inhibition of Human Coronaviruses by Combinations of Host-Targeted and Direct-Acting Antivirals. Antimicrobial Agents and Chemotherapy. 2023;67(4) [PubMed] | [CrossRef] | [Google Scholar]
- Feng H, Lakshmi KV, Ng CT, Sasikumar M, Arip M, Hassan M, et al. Update on Remdesivir in the Treatment of Novel Coronavirus Pneumonia. Progress In Microbes and Molecular Biology. 2023;6(1) [CrossRef] | [Google Scholar]
- Bege M, Borbás A. The Design, Synthesis and Mechanism of Action of Paxlovid, a Protease Inhibitor Drug Combination for the Treatment of COVID-19. Pharmaceutics. 2024;16(2):217 [PubMed] | [CrossRef] | [Google Scholar]
- Bekheit MS, Panda SS, Girgis A. Potential RNA-dependent RNA polymerase (RdRp) inhibitors as prospective drug candidates for SARS-CoV-2. European Journal of Medicinal Chemistry. 2023;252:115292 [PubMed] | [CrossRef] | [Google Scholar]
- Siripongboonsitti T, Muadchimkaew M, Tawinprai K, Issaranon O, Meepholkij W, Arttawejkul P, et al. Favipiravir treatment in non-severe COVID-19: promising results from multicenter propensity score-matched study (FAVICOV). Scientific Reports. 2023;13(1):14884 [PubMed] | [CrossRef] | [Google Scholar]
- Korula P, Alexander H, John JS, Kirubakaran R, Singh B, Tharyan P, Rupali P, et al. Favipiravir for treating COVID-19. Cochrane Database of Systematic Reviews. 2024;2(2) [PubMed] | [CrossRef] | [Google Scholar]
- Ruggeri M, Signorini A, Caravaggio S. Casirivimab and imdevimab: Cost-effectiveness analysis of the treatment based on monoclonal antibodies on outpatients with Covid-19. PLoS One. 2023;18(2) [PubMed] | [CrossRef] | [Google Scholar]
- Somersan-Karakaya S, Mylonakis E, Menon VP, Wells JC, Ali S, Sivapalasingam S, et al. Casirivimab and imdevimab for the treatment of hospitalized patients with COVID-19. The Journal of Infectious Diseases. 2023;227(1):23-34. [PubMed] | [CrossRef] | [Google Scholar]
- Wicaksono IA, Suhandi C, Elamin KM, Wathoni N. Efficacy and safety of casirivimab-imdevimab combination on COVID-19 patients: A systematic review and meta-analysis randomized controlled trial. Heliyon. 2023;9(12) [PubMed] | [CrossRef] | [Google Scholar]
- Morsali M, Doosti-Irani A, Amini S, Nazemipour M, Mansournia MA, Aliannejad R, et al. Comparison of corticosteroids types, dexamethasone and methylprednisolone in patients hospitalized with COVID-19: A systematic review and network meta-analysis. Global Epidemiology. 2023:100116 [PubMed] | [CrossRef] | [Google Scholar]
- Bain CR, Myles PS, Corcoran T, Dieleman JM. Postoperative systemic inflammatory dysregulation and corticosteroids: a narrative review. Anaesthesia. 2023;78(3):356-70. [PubMed] | [CrossRef] | [Google Scholar]
- Ahmed MS, Farag AB, Boys IN, Wang P, Menendez-Montes I, Nguyen NUN, et al. FDA approved drugs with antiviral activity against SARS-CoV-2: From structure-based repurposing to host-specific mechanisms. Biomedicine and Pharmacotherapy. 2023;162:114614 [PubMed] | [CrossRef] | [Google Scholar]
- Madhav H, Rehan M, Hoda N, Jameel E. Ivermectin as a Repurposed Drug for COVID-19 Publisher. 2023:47-68. [CrossRef] | [Google Scholar]
- Jitobaom K, Boonarkart C, Manopwisedjaroen S, Punyadee N, Borwornpinyo S, Thitithanyanont A, et al. Favipiravir and ivermectin show synergistic antiviral activity against SARS-CoV-2. Acta Virologica. 2023;67:12265 [CrossRef] | [Google Scholar]
- Miatmoko A, Yulistiani Sulistyowati MI, Setyawan D, Cahyani DM. The Use of Azithromycin and Lopinavir-Ritonavir in the Treatment of COVID-19, in Viral Drug Delivery Systems: Advances in Treatment of Infectious Diseases. 2023:339-60. [CrossRef] | [Google Scholar]
- Razmi M, Hashemi F, Gheytanchi E, Manshadi MD, Ghods R, Madjd Z, et al. Immunomodulatory-based therapy as a potential promising treatment strategy against severe COVID-19 patients: A systematic review. International Immunopharmacology. 2020;88:106942 [PubMed] | [CrossRef] | [Google Scholar]
- Snow TAC, Arulkumaran N, Singer M, Sang-Ho C. Effect of dexamethasone dose on outcomes in acute COVID-19 disease: A systematic review and meta-analysis. Journal of Infection. 2023;87(6):490-7. [PubMed] | [Google Scholar]
- Chevret S, Bouadma L, Dupuis C, Burdet C, Timsit J. Which severe COVID-19 patients could benefit from high dose dexamethasone? A Bayesian post-hoc reanalysis of the COVIDICUS randomized clinical trial. Annals of Intensive Care. 2023;13(1):75 [PubMed] | [CrossRef] | [Google Scholar]
- Horby PW, Mafham M, Peto L, Campbell M, Pessoa-Amorim G, Spata E, et al. Casirivimab and imdevimab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2022;399(10325):665-76. [PubMed] | [CrossRef] | [Google Scholar]
- Bierle DM, Ganesh R, Tulledge-Scheitel S, Hanson SN, Arndt LL, Wilker CG, Razonable R, et al. Monoclonal antibody treatment of breakthrough COVID-19 in fully vaccinated individuals with high-risk comorbidities. The Journal of Infectious Diseases. 2022;225(4):598-602. [PubMed] | [CrossRef] | [Google Scholar]
- Janssen NA, Grondman I, de Nooijer AH, Boahen CK, Koeken VA, Matzaraki V, et al. Dysregulated innate and adaptive immune responses discriminate disease severity in COVID-19. The Journal of Infectious Diseases. 2021;223(8):1322-33. [PubMed] | [CrossRef] | [Google Scholar]
- Wang Y, Luu LDW, Liu S, Zhu X, Huang S, Li F, et al. Single-cell transcriptomic analysis reveals systemic immune dysregulation in COVID-19-associated pediatric encephalopathy. Signal Transduction and Targeted Therapy. 2023;8(1):398 [PubMed] | [CrossRef] | [Google Scholar]
- Yin K, Peluso MJ, Luo X, Thomas R, Shin M-G, Neidleman J, et al. Long COVID manifests with T cell dysregulation, inflammation and an uncoordinated adaptive immune response to SARS-CoV-2. Nature Immunology. 2024;25(2):218-25. [PubMed] | [CrossRef] | [Google Scholar]
- Sebbar E-h, Choukri M. Interleukin 6: A biomarker for COVID-19 progression. Materials Today: Proceedings. 2023;72:3351-55. [PubMed] | [CrossRef] | [Google Scholar]
- Farhan NH, Abidalmutalib Aljabawi R. Tocilizumab for severe COVID-19: Review and meta-analysis. International Journal of Antimicrobial Agents. 2023;56(3):106103 [PubMed] | [CrossRef] | [Google Scholar]
- Al Kharusi M, Al Sheikh N, Alhajri M, Al Mandhri SA, Khafagy E-S, Moglad EH, et al. A Prospective Cohort Study of COVID-19: Evaluation of the Early Role of IL-1 and IL-6 Antagonists in Improving the Outcome of the Illness and Reduction in the Risk of Death. Healthcare. 2023;11(7):1025 [PubMed] | [CrossRef] | [Google Scholar]
- Pal R, Chaudhary MJ. Pharmacotherapeutics for cytokine storm in COVID-19, Stem Cells. 2024:101-125. [PubMed] | [CrossRef] | [Google Scholar]
- Mohammed MA. Fighting cytokine storm and immunomodulatory deficiency: By using natural products therapy up to now. Frontiers in Pharmacology. 2023;14:1111329 [PubMed] | [CrossRef] | [Google Scholar]
- Lu D-Y, Lu T-R. COVID-19 Study, Diagnostic and Therapeutic Transition. Recent Advances in Anti-Infective Drug Discovery. 2024;19(1):21-35. [PubMed] | [CrossRef] | [Google Scholar]
- Greenblatt W, Gupta C, Kao J. Drug Repurposing During The COVID-19 Pandemic: Lessons For Expediting Drug Development And Access: Study examines drug repurposing during the COVID-19 Pandemic and offers lessons learned for for both future emerging diseases and drug development in general. Health Aff. 2023;42(3):424-32. [PubMed] | [CrossRef] | [Google Scholar]
- Grobler JA, anderson AS, Fernandes P, Diamond MS, Colvis CM, Menetski JP, Alvarez RM, Young JAT, Carter KL, et al. Accelerated Preclinical Paths to Support Rapid Development of COVID-19 Therapeutics Cell Host and Microbe. 2020;28(5):638-45. [PubMed] | [CrossRef] | [Google Scholar]
- Tarique M, Ahmad S, Malik A, Ahmad I, Saeed M, Almatroudi A, Qadah T, Abdulaziz Murad M, Mashraqi M, Alam Q, Al-Saleh Y, et al. Novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) and other coronaviruses: a genome-wide comparative annotation and analysis. Molecular and Cellular Biochemistry. 2022;477(2):645 [PubMed] | [CrossRef] | [Google Scholar]
- Oliveira AM, Rudrapal M. Challenges and Regulatory Issues in Drug Repurposing and Computational Drug Discovery, in Drug Repurposing and Computational Drug Discovery. 2024:243-65. [PubMed] | [CrossRef] | [Google Scholar]
- Khaja UM, Bhat AH, Ahmed M, Ali A, Ganie SA. Pharmacogenomics in viral diseases, in Pharmacogenomics. 2023:247-69. [PubMed] | [CrossRef] | [Google Scholar]
- Abulsoud AI, El-Husseiny HM, El-Husseiny AA, El-Mahdy HA, Ismail A, Elkhawaga SY, et al. Mutations in SARS-CoV-2: Insights on structure, variants, vaccines and biomedical interventions. Biomedicine and Pharmacotherapy. 2023;157:113977 [PubMed] | [CrossRef] | [Google Scholar]
- Chakraborty C, Chatterjee S, Bhattacharya M, Chopra H, Bhattacharya P, Islam MA, Dhama K, et al. The D614G mutation helps to increase the transmissibility and reduce the virulence of SARS-CoV-2 variants through natural selection. International Journal of Surgery. 2023;109(2):171-4. [PubMed] | [CrossRef] | [Google Scholar]
- Alzahrani A, Alharbi S, Almitairi J, Alzahrani A, Alshehri MA, Aldughaim MS, Alothaid H, et al. Effectiveness of Early Favipiravir Therapy in Hospitalised COVID-19 Patients. Advances in Virology. 2022:9240941 [PubMed] | [CrossRef] | [Google Scholar]
- Ati EM, Abdulmajeed AM, Alharbi BM, Ajmi RM, Latif AS. Traceability environmental effects of microfabric in leaves of Cupressus dupreziana plant and Soil Surrounding it given the rise in COVID19. Advancements in Life Sciences. 2023;10(4):663-9. [PubMed] | [CrossRef] | [Google Scholar]
- Sadri A. Is Target-Based Drug Discovery Efficient? Discovery and “Off-Target” Mechanisms of All Drugs. Journal of Medicinal Chemistry. 2023;66(18):12651-77. [PubMed] | [CrossRef] | [Google Scholar]
- Burgess T, Rennie S, Moodley K. Key ethical issues encountered during COVID-19 research: a thematic analysis of perspectives from South African research ethics committees. BMC Medical Ethics. 2023;24(1):11 [PubMed] | [CrossRef] | [Google Scholar]
- Organization WH. Access to NCD medicines: emergent issues during the COVID-19 pandemic and key structural factors. World Health Organization. 2023:172 [PubMed] | [CrossRef] | [Google Scholar]
- Reynolds L, Dewey C, Asfour G, Little M. Vaccine efficacy against SARS-CoV-2 for Pfizer BioNTech, Moderna and AstraZeneca vaccines: a systematic review. Frontiers in Public Health. 2023:11 [PubMed] | [CrossRef] | [Google Scholar]
- Soheili M, Khateri S, Moradpour F, Mohammadzedeh P, Zareie M, Mortazavi S, et al. The efficacy and effectiveness of COVID-19 vaccines around the world: a mini-review and meta-analysis. Annals of Clinical Microbiology and Antimicrobials. 2023;22(1):1-14. [PubMed] | [CrossRef] | [Google Scholar]
- Singh P, Dhalaria P, Kashyap S, Soni GK, Nandi P, Ghosh S, et al. Strategies to overcome vaccine hesitancy: a systematic review. Systematic Reviews. 2022;11(1):1-13. [PubMed] | [CrossRef] | [Google Scholar]
- Chavda VP, Bezbaruah R, Valu D, Patel B, Kumar A, Prasad S, et al. Adenoviral vector-based vaccine platform for COVID-19: Current status. Vaccines. 2023;11(2):432 [PubMed] | [CrossRef] | [Google Scholar]
- Lundstrom KJ. Viral vector-based vaccines against SARS-CoV-2. Exploration of Immunology. 2021;1(4):295-308. [CrossRef] | [Google Scholar]
- Livingston EH, Malani PN, Creech CB. The Johnson and Johnson Vaccine for COVID-19. JAMA. 2021;325(15):1575 [PubMed] | [CrossRef] | [Google Scholar]
- Haider T, Ali J, Ali SM, Iftikhar AS, Siddiqui AA, Khan AS, et al. Prevalence of side effects of the AstraZeneca COVID-19 vaccine: a multicenter experience from Pakistan. Cureus. 2023;15(10) [PubMed] | [CrossRef] | [Google Scholar]
- McCann N, O’Connor D, Lambe T, Pollard A. Viral vector vaccines. Current Opinion in Immunology. 2022;77:102210 [PubMed] | [CrossRef] | [Google Scholar]
- Hayawi K, Shahriar S, Serhani MA, Alashwal H, Masud MM. Vaccine versus variants (3Vs): are the COVID-19 vaccines effective against the variants? A systematic review. Vaccines. 2021;9(11):1305 [PubMed] | [CrossRef] | [Google Scholar]
- Fiolet T, Kherabi Y, MacDonald C-J, Ghosn J, Peiffer-Smadja N. Comparing COVID-19 vaccines for their characteristics, efficacy and effectiveness against SARS-CoV-2 and variants of concern: a narrative review. Clinical Microbiology and Infection. 2022;28(2):202-21. [PubMed] | [CrossRef] | [Google Scholar]
- Garg I, Sheikh AB, Pal S, Shekhar R. Mix-and-match COVID-19 vaccinations (heterologous boost): a review. Infectious Disease Reports. 2022;14(4):537-46. [PubMed] | [CrossRef] | [Google Scholar]
- Fahrni ML, Ismail IA-N, Refi DM, Almeman A, Yaakob NC, Saman KM, et al. Management of COVID-19 vaccines cold chain logistics: a scoping review. Journal of Pharmaceutical Policy and Practice. 2022;15(1):16 [PubMed] | [CrossRef] | [Google Scholar]
- D’Agostino V, Caranci F, Negro A, Piscitelli V, Tuccillo B, Fasano F, et al. A rare case of cerebral venous thrombosis and disseminated intravascular coagulation temporally associated to the COVID-19 vaccine administration. Journal of Personalized Medicine. 2021;11(4):285 [PubMed] | [CrossRef] | [Google Scholar]
- Momenzadeh M, Moayedi E, Dehghani SL, Shadabi S, Derakhshan Barjoei M, Mousavi MS, Pourrezaei S, et al. Possibility of the blood clot, thrombotic thrombocytopenia following injection of COVID-19-vaccine AstraZeneca; a systematic review. Journal of Preventive Epidemiology. 2023;8(2) [CrossRef] | [Google Scholar]
- Lupu D, Tiganasu R. COVID-19 vaccination and governance in the case of low, middle and high-income countries. BMC Public Health. 2023;23(1):1073 [PubMed] | [CrossRef] | [Google Scholar]
- Kochakkashani F, Kayvanfar V, Haji A. Supply chain planning of vaccine and pharmaceutical clusters under uncertainty: The case of COVID-19. Socio-Economic Planning Sciences. 2023;87:101602 [PubMed] | [CrossRef] | [Google Scholar]
- Privor-Dumm L, Excler J-L, Gilbert S, Karim SSA, Hotez PJ, Thompson D, Kim J, et al. Vaccine access, equity and justice: COVID-19 vaccines and vaccination. BMJ Global Health. 2023;8(6) [PubMed] | [CrossRef] | [Google Scholar]
- Haldane V, Ariyarajah A, Berry I, Loutet M, Salamanca-Buentello F, Upshur REG, et al. Global inequity creates local insufficiency: A qualitative study of COVID-19 vaccine implementation challenges in low-and-middle-income countries. PLoS One. 2023;18(2) [PubMed] | [CrossRef] | [Google Scholar]
- Storeng KT, de Bengy Puyvallée A, Stein F. COVAX and the rise of the ‘super public private partnership’for global health. Global Public Health. 2023;18(1):1987502 [PubMed] | [CrossRef] | [Google Scholar]
- Wong A, Barrero Guevara LA, Goult E, Briga M, Kramer SC, Kovacevic A, et al. The interactions of SARS-CoV-2 with cocirculating pathogens: Epidemiological implications and current knowledge gaps. PLOS Pathogens. 2023;19(3) [PubMed] | [CrossRef] | [Google Scholar]