Continuing Education Activity
Coronavirus disease 2019 (COVID-19), the highly contagious infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has had a catastrophic effect on the world’s demographics resulting in more than 2.9 million deaths worldwide, emerging as the most consequential global health crisis since the era of the influenza pandemic of 1918.
Objectives:
-
Identify the etiology and epidemiology of COVID-19.
-
Describe the clinical features and radiological findings expected in patients with COVID-19.
-
Summarize the latest available treatment in the management of COVID-19, including the different vaccines available to prevent COVID-19.
-
Discuss interprofessional team strategies for improving care coordination and communication to care for patients with coronavirus and improve outcomes.
Earn continuing education credits (CME/CE) on this topic.
Introduction
Coronavirus disease 2019 (COVID-19), the highly contagious infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has had a catastrophic effect on the world’s demographics resulting in more than 2.9 million deaths worldwide, emerging as the most consequential global health crisis since the era of the influenza pandemic of 1918. After the first cases of this predominantly respiratory viral illness were first reported in Wuhan, Hubei Province, China, in late December 2019, SARS-CoV-2 rapidly disseminated across the world in a short span of time, compelling the World Health Organization (WHO) to declare it as a global pandemic on March 11, 2020. Since being declared a global pandemic, COVID-19 has ravaged many countries worldwide, overwhelming many healthcare systems. The pandemic has also resulted in the loss of livelihoods due to prolonged shutdowns, which have had a rippling effect on the global economy. Even though substantial progress in clinical research has led to a better understanding of SARS-CoV-2 and the management of COVID-19, limiting the continuing spread of this virus has become an issue of increasing concern, as SARS-CoV-2 continues to wreak havoc across the world, with many countries enduring a second or third wave of outbreaks of this viral illness attributed mainly due to the emergence of mutant variants of the virus.
Like other RNA viruses, SARS-CoV-2, while adapting to their new human hosts, is prone to genetic evolution with the development of mutations over time, resulting in variants that may have different characteristics than its ancestral strains. Several variants of SARS-CoV-2 have been described during the course of this pandemic, among which only a few are considered variants of concern (VOCs), given their impact on public health. The B.1.1.7 lineage (or VOC 202012) variant was the first VOC described in the United Kingdom (UK) in late December 2020, and subsequently, the B.1.351 lineage (or 501Y.V2) was reported in South Africa. A third VOC, B.1.1.248/B1.1.28/P1 (or 501Y.V3), was reported in Brazil in early January 2021, and more recently, the B.1.427/B.1.429 lineage was identified in California. Despite the unprecedented speed of vaccine development against the prevention of COVID-19 and robust global mass vaccination efforts, the emergence of these new SARS-CoV-2 variants threatens to overturn the significant progress made so far in limiting the transmission of this virus.
This review article aims to comprehensively describe the etiology, epidemiology, pathophysiology, clinical features, diagnostic methods, and the latest novel therapeutics in the management of COVID-19. This review also briefly provides an overview of the different variants of SARS-CoV-2 and the efficacy of different available vaccines for prevention against COVID-19 and its variants.
Etiology
Coronaviruses (CoVs) are positive-stranded RNA(+ssRNA) viruses with a crown-like appearance under an electron microscope (coronam is the Latin term for crown) due to the presence of spike glycoproteins on the envelope. The subfamily Orthocoronavirinae of the Coronaviridae family (order Nidovirales) classifies into four genera of CoVs:
-
Alphacoronavirus (alphaCoV)
-
Betacoronavirus (betaCoV)
-
Deltacoronavirus (deltaCoV)
-
Gammacoronavirus (gammaCoV)
BetaCoV genus is further divided into five sub-genera or lineages.[1] Genomic characterization has shown that bats and rodents are the probable gene sources of alphaCoVs and betaCoVs. On the contrary, avian species seem to represent the gene sources of deltaCoVs and gammaCoVs. CoVs have become the major pathogens of emerging respiratory disease outbreaks. Members of this large family of viruses can cause respiratory, enteric, hepatic, and neurological diseases in different animal species, including camels, cattle, cats, and bats. For reasons yet to be explained, these viruses can cross species barriers and can cause, in humans, illness ranging from the common cold to more severe diseases such as MERS and SARS. To date, seven human CoVs (HCoVs) capable of infecting humans have been identified. Some of HCoVs were identified in the mid-1960s, while others were only detected in the new millennium. In general, estimates suggest that 2% of the population are healthy carriers of a CoVs and that these viruses are responsible for about 5% to 10% of acute respiratory infections.[2]
-
Common human CoVs: HCoV-OC43, and HCoV-HKU1 (betaCoVs of the A lineage); HCoV-229E, and HCoV-NL63 (alphaCoVs). These viruses can cause common colds and self-limiting upper respiratory tract infections in immunocompetent individuals. However, in immunocompromised subjects and the elderly, lower respiratory tract infections can occur due to these viruses.
-
Other human CoVs: SARS-CoV and MERS-CoV (betaCoVs of the B and C lineage, respectively). These viruses are considered to be more virulent and capable of causing epidemics manifesting with respiratory and extra-respiratory manifestations of variable clinical severity.
SARS-CoV-2 is a novel betaCoV belonging to the same subgenus as the severe acute respiratory syndrome coronavirus (SARS-CoV) and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV), which have been previously implicated in SARS-CoV and MERS-CoV epidemics with mortality rates up to 10% and 35%, respectively.[3] It has a round or elliptic and often pleomorphic form and a diameter of approximately 60–140 nm. Like other CoVs, it is sensitive to ultraviolet rays and heat. In this regard, although high temperature decreases the replication of any species of virus. Currently, the inactivation temperature of SARS-CoV-2 must be well elucidated. It appears that this virus can be inactivated at about 27° C. Conversely, it may resist lower temperatures even below 0°C. Also, these viruses can be effectively inactivated by lipid solvents, including ether (75%), ethanol, chlorine-containing disinfectant, peroxyacetic acid, and chloroform except for chlorhexidine.
Genomic characterization of the new HCoV, isolated from a cluster-patient with atypical pneumonia after visiting Wuhan, had 89% nucleotide identity with bat SARS-like-CoVZXC21 and 82% with that of human SARS-CoV. Hence, it was termed SARS-CoV-2 by experts of the International Committee on Taxonomy of Viruses. The single-stranded RNA genome of SARS-CoV-2 contains 29891 nucleotides, encoding for 9860 amino acids.
Although the origin of SARS-CoV-2 is currently unknown, it is widely postulated to have originated from an animal implicating zoonotic transmission. Genomic analyses suggest that SARS-CoV-2 probably evolved from a strain found in bats. The genomic comparison between the human SARS-CoV-2 sequence and known animal coronaviruses indeed revealed high homology (96%) between the SARS-CoV-2 and the betaCoV RaTG13 of bats (Rhinolophus affinis)[4]Similar to SARS and MERS, it has been hypothesized that SARS-CoV-2 advanced from bats to intermediate hosts such as pangolins and minks, and then to humans.[5][6] A recently released report by the WHO describing the possible origins of SARS-CoV-2 was inconclusive as it did not clearly specify the origin of the virus; however, it did report that the circulation of SARS-CoV-2 occurred as early as December 2019. This report explored several possible hypotheses of the origin of the virus that included the origin of the virus in an animal, the transmission of the virus to an intermediate host, and subsequent passage into humans.
SARS-CoV-2 Variants
As mentioned earlier, SARS-CoV-2 is prone to genetic evolution resulting in multiple variants that may have different characteristics compared to its ancestral strains. Periodic genomic sequencing of viral samples is of fundamental importance, especially in a global pandemic setting, as it helps detect any new genetic variants of SARS-CoV-2. Notably, the genetic evolution was minimal initially with the emergence of the globally dominant D614G variant, which was associated with increased transmissibility but without the ability to cause severe illness.[7] Another variant was identified in humans, attributed to transmission from infected farmed mink in Denmark, which was not associated with increased transmissibility[6]. Since then, multiple variants of SARS-CoV-2 have been described, of which a few are considered variants of concern (VOCs) due to their potential to cause enhanced transmissibility or virulence, reduction in neutralization by antibodies obtained through natural infection or vaccination, the ability to evade detection, or a decrease in therapeutics or vaccination effectiveness. With the continued emergence of multiple variants, the CDC and the WHO have independently established a classification system for distinguishing the emerging variants of SARS-CoV-2 into variants of concern(VOCs) and variants of interest(VOIs).
SARS-CoV-2 Variants of Concern (VOCs)
-
501Y.V2 (B.1.351 lineage)
-
- A new variant of SARS-CoV-2 lineage (B.1.351 lineage or 501Y.V2) with multiple spike mutations, which resulted in the second wave of COVID-19 infections, was first detected in South Africa in October 2020.[17]
- The B.1.351 variant includes nine mutations (L18F, D80A, D215G, R246I, K417N, E484K, N501Y, D614G, and A701V) in the spike protein, of which three mutations (K417N, E484K, and N501Y) are located in the RBD and increase the binding affinity for the ACE receptors.[18][18][10][19] SARS-CoV-2 501Y.V2(B.1.351 lineage) was reported in the US at the end of January 2021.
- This variant is reported to have an increased risk of transmission and reduced neutralization by monoclonal antibody therapy, convalescent sera, and post-vaccination sera.[20]
-
P.1 (B.1.1.28.1 lineage)
-
- The third variant of concern, the B.1.1.28 variant, also known as 501Y.V3 or P.1 lineage, was identified in December 2020 in Brazil and was first detected in the US in January 2021.[21]
- The B.1.1.28 variant harbors ten mutations in the spike protein (L18F, T20N, P26S, D138Y, R190S, H655Y, T1027I V1176, K417T, E484K, and N501Y). Three mutations (L18F, K417N, E484K) are located in the RBD, similar to the B.1.351 variant.[21]
- Notably, this variant may have reduced neutralization by monoclonal antibody therapies, convalescent sera, and post-vaccination sera.[20]
SARS-CoV-2 Variants of Interest (VOI)
VOIs are defined as variants with specific genetic markers that have been associated with changes that may cause enhanced transmissibility or virulence, reduction in neutralization by antibodies obtained through natural infection or vaccination, the ability to evade detection, or a decrease in the effectiveness of therapeutics or vaccination. The WHO Epidemiological update April 13, 2021, described seven variants of interest (VOIs), namely B.1.427/B.1.429; B.1.525; B.1.526; B.1.1.28.2 alias P2 ;B.1.1.28.3, alias P.3 and B.1.616. The CDC also has described three VOIs, B.1.525, B.1.526, and B.1.1.28.2.
- SARS-CoV-2 B.1.427 and B.1.429 variants, also called CAL.20C/L452R, emerged in the US around June 2020 and increased from 0% to >50% of sequenced cases from September 1, 2020, to January 29, 2021, exhibiting an 18.6-24% increase in transmissibility relative to wild-type circulating strains. These variants harbor specific mutations (B.1.427: L452R, D614G; B.1.429: S13I, W152C, L452R, D614G). Due to its increased transmissibility, the CDC classified this strain as a variant of concern in the US.[22]
-
SARS-CoV-2 B.1.525 and B.1.526 harbor key spike mutations (B.1.525: A67V, Δ69/70, Δ144, E484K, D614G, Q677H, F888L; B.1.526: (L5F*), T95I, D253G, (S477N*), (E484K*), D614G, (A701V*)) and were first detected in New York in November 2020 and classified as a variant of interest by CDC and the WHO due to their potential reduction in neutralization by antibody treatments and vaccine sera.
-
SARS-CoV-2 B.1.1.28.3/P3 variant, also called PHL-B.1.1.28, has key spike mutations (141-143 deletion E484K; N501Y; and P681H) and was first detected in the Philippines and Japan in February 2021 and is classified as a variant of interest by the WHO.
Transmission of SARS-CoV-2
-
The primary mode of transmission of SARS-CoV-2 is via exposure to respiratory droplets carrying the infectious virus from close contact or droplet transmission from presymptomatic, asymptomatic, or symptomatic individuals harboring the virus.
-
Airborne transmission with aerosol-generating procedures has also been implicated in the spread of COVID-19. However, data implicating airborne transmission of SARS-CoV-2 is emerging but has not been universally acknowledged.
-
Fomite transmission from contamination of inanimate surfaces with SARS-CoV-2 has been well characterized based on many studies reporting the viability of SARS-CoV-2 on various porous and nonporous surfaces.
- Under experimental conditions, SARS-CoV-2 was noted to be stable on stainless steel and plastic surfaces compared to copper and cardboard surfaces, with the viable virus being detected up to 72 hours after inoculating the surfaces with the virus.[23]
- Viable virus was isolated for up to 28 days at 20 degrees C from nonporous surfaces such as glass, stainless steel. Conversely, recovery of SARS-CoV-2 on porous materials was reduced compared with nonporous surfaces.[24]
- A study evaluating the duration of the viability of the virus on objects and surfaces showed that SARS-CoV-2 can be found on plastic and stainless steel for up to 2-3 days, cardboard for up to 1 day, copper for up to 4 hours. Moreover, it seems that contamination was higher in intensive care units (ICUs) than in general wards, and SARS-CoV-2 can be found on floors, computer mice, trash cans, and sickbed handrails as well as in the air up to 4 meters from patients implicating nosocomial transmission as well in addition to fomite transmission.[25]
-
The Centers for Disease Control and Prevention(CDC) recently released an update stating that individuals can be infected with SARS-CoV-2 via contact with surfaces contaminated by the virus, but the risk is low and is not the main route of transmission of this virus.
- Epidemiologic data from several case studies have reported that patients with SARS-CoV-2 infection have the live virus present in feces implying possible fecal-oral transmission.[26]
- A meta-analysis that included 936 neonates from mothers with COVID-19 showed vertical transmission is possible but occurs in a minority of cases.[27]
Epidemiology
According to the World Health Organization (WHO), the emergence of viral diseases represents a serious public health risk. In the past two decades, several epidemics caused by viruses such as the severe acute respiratory syndrome coronavirus (SARS-CoV) from 2002 to 2003, and H1N1 influenza in 2009, and the Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012 have been described which have had a significant impact on global health. Since being declared a global pandemic by the WHO, SARS-CoV-2, the virus responsible for COVID-19 has spread to 223 countries with more than 138 million cases, and more than 2.9 million deaths reported globally. The U.S. experienced the highest number of SARS-CoV-2 infections and COVID-19 related deaths. In fact, COVID-19 was the third leading cause of death in the U.S. in 2020 after heart disease and cancer, with approximately 375,000 death reported.[28] An epidemiological update by the WHO on April 13, 2021, reported that SARS-CoV-2 (B.1.1.7 lineage/ VOC 202012/01) has spread to 132 countries, the SARS-CoV-2 501Y.V2 (B.1.351 lineage) has been reported in 82 countries. The SARS-CoV-2 P.1 (B.1.1.28.1) has been detected in 52 countries around the world. The WHO’s current estimate of the global case fatality rate for COVID-19 is 2.2%. However, the case fatality rate is affected by factors that include age, underlying preexisting conditions, and severity of illness and significantly varies between countries.
Age, Gender-based Differences And The Impact Of Medical Comorbidities in COVID-19
Individuals of all ages are at risk of contracting this infection and severe disease. However, patients aged ≥60 years and patients with underlying medical comorbidities (obesity, cardiovascular disease, chronic kidney disease, diabetes, chronic lung disease, smoking, cancer, solid organ or hematopoietic stem cell transplant patients) have an increased risk of developing severe COVID-19 infection. The percentage of COVID-19 patients requiring hospitalization was six times higher in those with preexisting medical conditions than those without medical conditions (45.4% vs. 7.6%) based on an analysis by Stokes et al. of confirmed cases reported to the CDC during January 22 to May 30, 2020. Notably, the study also reported that the percentage of patients who succumbed to this illness was 12 times higher in those with preexisting medical conditions than those without medical conditions (19.5% vs. 1.6%).[29] Data regarding the gender-based differences in COVID-19 suggests that male patients are at risk of developing severe illness and increased mortality due to COVID-19 compared to female patients.[30][31] Results from a retrospective cohort study from March 1 to November 21, 2020, evaluating the mortality rate in 209 US acute care hospitals that included 42 604 patients with confirmed SARS-CoV-2 infection, reported a higher mortality rate in male patients (12.5%) compared to female patients (9.6%).[32]
Racial and Ethnic Disparities in COVID-19
The severity of infection and mortality related to COVID-19 also varies between different ethnic groups.[30] Racial and ethnic minority groups were reported to have a higher percentage of COVID-19 related hospitalizations than White patients based on a recent CDC analysis of hospitalizations from a large administrative database that included approximately 300,000 COVID-19 patients hospitalized from March 2020 to December 2020. This high percentage of COVID-19 related hospitalizations among racial and ethnic groups was driven by a higher risk for exposure to SARS-CoV-2 and an increased risk for developing severe COVID-19 disease.[33] The results of a meta-analysis of 50 studies from the US and UK researchers noted that people of Black, Hispanic, and Asian ethnic minority groups are at increased risk of contracting and dying from COVID-19 infection.[34] COVID-19 related death rates were the highest among Hispanic persons.[28] Another analysis by the CDC evaluating the risk of COVID-19 among sexual minority adults reported that underlying medical comorbidities which increase the risk of developing severe COVID-19 were more prevalent in sexual minority individuals than heterosexual individuals both within the general population and within specific racial/ethnic groups.[35]
Pathophysiology
The general description of viral structure and its genome of CoVs is essential for addressing the pathogenesis of SARS-CoV-2. As described earlier, CoVs are enveloped, positive-stranded RNA viruses with a nucleocapsid, and the genomic structure is organized in a +ssRNA of approximately 30 kb in length and with a 5′-cap structure and 3′-poly-A tail making it the largest among RNA viruses. Upon entry into the host, replication of the viral RNA is initiated with the synthesis of polyprotein 1a/1ab (pp1a/pp1ab). The transcription occurs through the replication-transcription complex (RCT) organized in double-membrane vesicles and via the synthesis of subgenomic RNAs (sgRNAs) sequences. Conversely, transcription termination occurs at transcription regulatory sequences, located between the so-called open reading frames (ORFs) that work as templates for the production of subgenomic mRNAs. In an atypical CoV genome, at least six ORFs can be present. Among these, a frameshift between ORF1a and ORF1b guides the production of both pp1a and pp1ab polypeptides that are processed by virally encoded chymotrypsin-like protease (3CLpro) or main protease (Mpro), as well as one or two papain-like proteases for producing 16 with known or predicted RNA synthesis and modification functions non-structural proteins (NSPs 1-16). Besides ORF1a and ORF1b, other ORFs encode structural proteins, including spike, membrane, envelope, and nucleocapsid proteins and accessory proteic chains.[3]. Different CoVs possess unique structural and accessory proteins translated by dedicated sgRNAs.The pathogenesis of CoVs and SARS-CoV-2 is related to the function of the NSPs and structural proteins. For example, researchers have outlined the role of NSPs in blocking the host\’s innate immune response.[2] Among functions of structural proteins, the envelope has a crucial role in virus pathogenicity as it promotes viral assembly and release. Among the structural elements of CoVs, there are the spike glycoproteins composed of two subunits (S1 and S2). Homotrimers of S proteins compose the spikes on the viral surface, guiding the link to host receptors.[36]
Pathogenesis of SARS-CoV-2
Structurally and phylogenetically, SARS-CoV-2 is similar to SARS-CoV and MERS-CoV and is composed of four main structural proteins: spike (S), envelope (E) glycoprotein, nucleocapsid (N), membrane (M) protein, along with 16 nonstructural proteins, and 5-8 accessory proteins.[37] The surface spike (S) glycoprotein, which resembles a crown, is located on the outer surface of the virion and undergoes cleavage into an amino (N)-terminal S1 subunit, which facilitates the incorporation of the virus into the host cell and a carboxyl (C)-terminal S2 subunit containing a fusion peptide, a transmembrane domain, and cytoplasmic domain is responsible for virus-cell membrane fusion.[38][39] The S1 subunit is further divided into a receptor-binding domain (RBD) and N-terminal domain (NTD), which facilitates viral entry into the host cell and serves as a potential target for neutralization in response to antisera or vaccines.[36] The RBD is a fundamental peptide domain in the pathogenesis of infection as it represents a binding site for the human angiotensin-converting enzyme 2 (ACE2) receptors. Inhibition of the renin-angiotensin-aldosterone system(RAAS), as previously hypothesized, does not increase the risk of hospitalization for COVID-19 and severe disease.[40]
SARS-CoV-2 gains entry into the hosts\’ cells by binding the SARS-CoV-2 spike or S protein (S1) to the ACE2 receptors abundantly on respiratory epithelium such as type II alveolar epithelial cells. Besides the respiratory epithelium, ACE2 receptors are also expressed by other organs such as the upper esophagus, enterocytes from the ileum, myocardial cells, proximal tubular cells of the kidney, and urothelial cells of the bladder.[41] The viral attachment process is followed by priming the spike protein S2 subunit by the host transmembrane serine protease 2 (TMPRSS2) that facilitates cell entry and subsequent viral replication endocytosis with the assembly of virions.[42]
In summary, the spike RBD allows the binding to the ACE2 receptor in the lungs and other tissues. The spike protein of an amino acid site (polybasic site) allows the functional processing of the same by the human enzyme furin (protease). This process enables the exposure of the fusion sequences and, therefore, the fusion of the viral and cell membranes, a necessary passage for the virus to enter the cell.
Effect of SARS-CoV-2 on the Respiratory System/Pathogenesis of SARS-CoV-2-induced Pneumonia
COVID-19 is primarily considered a viral respiratory illness as its causative agent, SARS-CoV-2, predominantly targets the respiratory system.
The pathogenesis of SARS-CoV-2 induced pneumonia is best explained by two stages, an early and a late phase. The early phase is characterized by viral replication resulting in direct virus-mediated tissue damage, which is followed by a late phase when the infected host cells trigger an immune response with the recruitment of T lymphocytes, monocytes, and neutrophil recruitment which releases cytokines such as tumor necrosis factor-α (TNF α), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-1 (IL-1), interleukin-6 (IL-6), ), IL-1β, IL-8, IL-12 and interferon (IFN)-γ. In severe COVID-19, the immune system\’s overactivation results in a \’cytokine storm\’ characterized by the release of high levels of cytokines, especially IL-6 and TNF-α, into the circulation, causing a local and systemic inflammatory response.[43][44] The increased vascular permeability and subsequent development of pulmonary edema in patients with severe COVID-19 are explained by multiple mechanisms, which includes a) endotheliitis as a result of direct viral injury and perivascular inflammation leading to microvascular and microthrombi deposition b) dysregulation of the RAAS due to increased binding of the virus to the ACE2 receptors and c)activation of the kallikrein- bradykinin pathway, the activation of which enhances vascular permeability, d)enhanced epithelial cell contraction causing swelling of cells and disturbance of intercellular junctions.[45][46][47] Besides IL-6 and TNF-α, binding of SARS-CoV-2 to the Toll-Like Receptor (TLR) induces the release of pro-IL-1β, which is cleaved into the active mature IL-1β that mediates lung inflammation, until fibrosis.[48]
Effect of SARS-CoV-2 on Extrapulmonary Organ Systems
Although the respiratory system is the principal target for SARS-CoV-2 as described above, it can affect other major organ systems such as the gastrointestinal tract (GI), hepatobiliary, cardiovascular, renal, and central nervous system. SARS-CoV-2–induced organ dysfunction, in general, is possibly explained by either one or a combination of the proposed mechanisms such as direct viral toxicity, ischemic injury caused by vasculitis, thrombosis, or thrombo-inflammation, immune dysregulation, and renin-angiotensin-aldosterone system (RAAS) dysregulation.[49]
- Cardiovascular system (CVS): Although the exact mechanism of cardiac involvement in COVID-19 is unknown, it is likely multifactorial. ACE2 receptors are also exhibited by myocardial cells implicating direct cytotoxicity by the SARS-CoV-2 on the myocardium leading to myocarditis. Proinflammatory cytokines such as IL-6 can also lead to vascular inflammation, myocarditis, and cardiac arrhythmias.[50] Acute coronary syndrome (ACS) a well-recognized cardiac manifestation of COVID-19 and is likely due to multiple factors that include but not limited to COVID-19 associated hypercoagulability, the release of proinflammatory cytokines, worsening of preexisting severe coronary artery disease, stress cardiomyopathy, and associated hemodynamic derangement which may reduce coronary blood flow, reduced oxygen supply resulting in the destabilization of coronary plaque microthrombogenesis or worsening of preexisting severe coronary artery disease.[51][52][53][54]
- Hematological: SARS-CoV-2 has a significant effect on the hematological and hemostatic system. The mechanism of leukopenia, one of the most common laboratory abnormalities encountered in COVID-19, is unknown. Several hypotheses have been postulated that include ACE 2 mediated lymphocyte destruction by direct invasion by the virus, lymphocyte apoptosis due to proinflammatory cytokines, and possible invasion of the virus of the lymphatic organs.[55] Thrombocytopenia is uncommon in COVID-19 and is likely due to multiple factors that include virus-mediated suppression of platelets, formation of autoantibodies, and activation of coagulation cascade that results in platelet consumption.[56] Thrombocytopenia and neutrophilia are considered a hallmark of severe illness.[49] Although it is well known that COVID-19 is associated with a state of hypercoagulability, the exact mechanisms that lead to the activation of the coagulation system are unknown and likely attributed to the cytokine-induced inflammatory response. The pathogenesis of this associated hypercoagulability is multifactorial and is probably induced by direct viral-mediated damage or cytokine-induced injury of the vascular endothelium leading to the activation of platelets, monocytes, and macrophages, increased expression of tissue factor, von Willebrand factor, and Factor VIII that results in the generation of thrombin and formation of fibrin clot formation.[56][57][56] Other mechanisms that have been proposed include possible mononuclear phagocytes induced prothrombotic sequelae, derangements in the renin-angiotensin system (RAS) pathways, complement-mediated microangiopathy.[56]
- Central Nervous System (CNS): There is emerging evidence of ACE2 receptors in human and mouse brains, implicating the potential infection of the brain by SARS-CoV-2.[58] The possible routes by which SARS-CoV-2 can invade the central nervous system are transsynaptic transfer across infected neurons via the olfactory nerve, vascular endothelial cell infection, or migration of leukocytes across the blood-brain barrier.[59]
- Gastrointestinal (GI) Tract: The pathogenesis of GI manifestations of COVID-19 is unknown and is likely considered to be multifactorial due to several potential mechanisms that include the direct ACE 2-mediated viral cytotoxicity of the intestinal mucosa, cytokine-induced inflammation, gut dysbiosis, and vascular abnormalities.[60]
-
Hepatobiliary: Although the pathogenesis of liver injury in COVID-19 patients is unknown, hepatic injury in COVID-19 is likely multifactorial and is explained by many mechanisms alone or in combination that includes ACE-2-mediated viral replication in the liver, direct virus-mediated damage, hypoxic or ischemic injury, immune-mediated inflammatory response, drug-induced liver injury (DILI), or worsening of preexisting liver disease.
- Renal: The pathogenesis of COVID-19 associated kidney injury is unknown and is likely multifactorial explained by a single or a combination of many factors such as direct cytotoxic injury from the virus, imbalance in the RAAS, associated cytokine-induced hyperinflammatory state, microvascular injury, and the prothrombotic state associated with COVID-19. Other factors such as associated hypovolemia, potential nephrotoxic agents, and nosocomial sepsis can also potentially contribute to kidney injury.[61]
Implications of New Variants of SARS-CoV-2 on the Pathogenesis of COVID-19
Genetic variation in the viral genes of SARS-CoV-2 can have implications in its pathogenesis, especially if it involves the RBD, which mediates viral entry into the host cells and is an essential target of vaccine sera monoclonal antibodies. All three reported VOCs (B.1.1.7; B.1.351; and P.1) have mutations in the RBD and the NTD, of which N501Y mutation located on the RBD is common to all variants and results in increased affinity of the spike protein to ACE 2 receptors enhancing the viral attachment and its subsequent entry into the host cells. Along with NBD, RBD serves as the dominant neutralization target and facilitates antibody production in response to antisera or vaccines.[62] Two recent preprint studies (not peer-reviewed) reported that a single mutation of N501Y alone increases the affinity between RBD and ACE2 approximately ten times more than the ancestral strain (N501-RBD). Interestingly the binding affinity of B.1.351 variant and P.1 variant with mutations N417/K848/Y501-RBD and ACE2 was much lower than that of N501Y-RBD and ACE2.[63][51]
Histopathology
Lungs: A multicenter analysis of lung tissue obtained during autopsies of patients who tested positive for COVID-19 demonstrated typical diffuse alveolar damage features in 87% of cases. Additionally, there was a frequent presence of type II pneumocyte hyperplasia, airway inflammation, and hyaline membranes in alveolar zones. Forty-two percent of patients were noted to have large vessel thrombi, platelet (CD61 positive), and/or fibrin microthrombi were present in 84% of cases.[64]
Brain: A single-center histopathological study of brain specimens obtained from 18 patients who succumbed to COVID-19 demonstrated acute hypoxic injury in all patients\’ cerebrum and cerebellum. Notably, no features of encephalitis or other specific brain changes were seen. Additionally, immunohistochemical analysis of brain tissue did not show cytoplasmic viral staining.[65]
Heart: Analysis of cardiac tissue from 39 autopsy cases of patients who tested positive for SARS-CoV-2 demonstrated the presence of SARS-CoV-2 viral genome within the myocardium.[66]
Kidney: Histopathology analysis of kidney specimens obtained from autopsies of 26 patients with confirmed COVID-19 demonstrated signs of diffuse proximal tubular injury with loss of brush border, non-isometric vacuolar degeneration, and necrosis. Additionally, electron microscopy showed clusters of coronavirus-like particles with spikes in the tubular epithelium and podocytes.[67]
GI Tract: Endoscopic specimens demonstrated positive staining of the viral nucleocapsid protein in the gastric, duodenal, and rectal epithelium cytoplasm. Numerous infiltrating plasma cells and lymphocytes with interstitial edema were seen in the lamina propria of the stomach, duodenum, and rectum.[68]
Liver: A prospective single-center clinicopathologic case series study involving the postmortem histopathological exam of major organs of 11 deceased patients with COVID-19 reported hepatic steatosis findings in all patients. The liver specimens of 73% of patients demonstrated chronic congestion. Different forms of hepatocyte necrosis were noted in 4 patients, and 70% showed nodular proliferation.[69]
History and Physical
Clinical Manifestations of COVID-19
- The median incubation period for SARS-CoV-2 is estimated to be 5.1 days, and the majority of patients will develop symptoms within 11.5 days of infection.[70]
-
The clinical spectrum of COVID-19 varies from asymptomatic or paucisymptomatic forms to clinical illness characterized by acute respiratory failure requiring mechanical ventilation, septic shock, and multiple organ failure.
- It is estimated that 17.9% to 33.3% of infected patients will remain asymptomatic.[71][72]
- Conversely, the vast majority of symptomatic patients commonly present with fever, cough, and shortness of breath and less commonly with a sore throat, anosmia, dysgeusia, anorexia, nausea, malaise, myalgias, and diarrhea. Stokes et al. reported that among 373,883 confirmed symptomatic COVID-19 cases in the US, 70% of them experienced fever, cough, shortness of breath, 36% reported myalgia, and 34% reported headache.[29]
- A large meta-analysis evaluating clinicopathological characteristics of 8697 patients with COVID-19 in China reported laboratory abnormalities that included lymphopenia (47.6%), elevated C-reactive protein levels (65.9%), elevated cardiac enzymes (49.4%), and abnormal liver function tests (26.4%).[58] Other laboratory abnormalities included leukopenia (23.5%), elevated D-dimer (20.4%), elevated erythrocyte sedimentation rate (20.4%), leukocytosis (9.9%), elevated procalcitonin (16.7%), and abnormal renal function (10.9%).[73]
- A meta-analysis of 212 published studies comprising of 281,461 individuals from 11 countries/regions reported that severe disease course was noted in about 23% with a mortality rate of about 6% in patients infected COVID-19.[74]
- The elevated neutrophil-to-lymphocyte ratio (NLR), derived NLR ratio (d-NLR) [neutrophil count divided by the result of WBC count minus neutrophil count], and the platelet-to-lymphocyte ratio is indicative of a cytokine-induced inflammatory storm.[75]
Based on the severity of presenting illness that includes clinical symptoms, laboratory and radiographic abnormalities, hemodynamics, and organ function. The National Institutes of Health (NIH) issued guidelines that classify COVID-19 into five distinct types.
-
Asymptomatic or Presymptomatic Infection: Individuals with positive SARS-CoV-2 test without any clinical symptoms consistent with COVID-19.
-
Mild illness: Individuals who have any symptoms of COVID-19 such as fever, cough, sore throat, malaise, headache, muscle pain, nausea, vomiting, diarrhea, anosmia, or dysgeusia but without shortness of breath or abnormal chest imaging
-
Moderate illness: Individuals who have clinical symptoms or radiologic evidence of lower respiratory tract disease and who have oxygen saturation (SpO2) ≥ 94% on room air
-
Severe illness: Individuals who have (SpO2) ≤ 94% on room air; a ratio of partial pressure of arterial oxygen to fraction of inspired oxygen, (PaO2/FiO2) 30 breaths/min or lung infiltrates >50%.
-
Critical illness: Individuals who have acute respiratory failure, septic shock, and/or multiple organ dysfunction. Patients with severe COVID-19 illness may become critically ill with the development of acute respiratory distress syndrome (ARDS) that tends to occur approximately one week after the onset of symptoms.
ARDS is characterized by a severe new-onset respiratory failure or worsening of an already identified respiratory picture. The diagnosis requires a set of clinical and ventilatory criteria such as chest imaging utilized includes chest radiograph, CT scan, or lung ultrasound demonstrating bilateral opacities (lung infiltrates > 50%), not fully explained by effusions, lobar, or lung collapse. If there are clinical and radiologic findings of pulmonary edema, heart failure, or other causes such as fluid overload, they should be excluded before assessing it to be ARDS. The Berlin definition classifies ARDS into three types based on the degree of hypoxia, with the reference parameter being PaO2/FiO2 or P/F ratio[76]:
-
Mild ARDS: 200 mmHg < PaO2/FiO2 ≤ 300 mmHg in patients not receiving mechanical ventilation or in those managed through non-invasive ventilation (NIV) by using positive end-expiratory pressure (PEEP) or a continuous positive airway pressure (CPAP) ≥ 5 cmH2O.
-
Moderate ARDS: 100 mmHg < PaO2/FiO2 ≤ 200 mmHg
-
Severe ARDS: PaO2/FiO2 ≤ 100 mmHg.
When PaO2 is not available, a ratio of SpO2/FiO2 ≤ 315 is suggestive of ARDS. A multicenter prospective observational study that analyzed 28-day mortality in mechanically ventilated patients with ARDS concluded that COVID-19 ARDS patients had similar ARDS features from other causes. The risk of 28-day mortality increased with ARDS severity.[77]
Extrapulmonary Manifestations
Although COVID-19, the illness caused by SARS-CoV-2, predominantly affects the respiratory system, COVID-19 can be considered a systemic viral illness given the multiple organ dysfunction associated with this illness.
- Renal manifestations: Patients hospitalized with severe COVID-19 are at risk for developing kidney injury, most commonly manifesting as acute kidney injury (AKI), which is likely multifactorial in the setting of hypervolemia, drug injury, vascular injury, and drug-related injury, and possibly direct cytotoxicity of the virus itself. AKI is the most frequently encountered extrapulmonary manifestation of COVID-19 and is associated with an increased risk of mortality.[78] A large multicenter cohort study of hospitalized patients with COVID-19 that involved 5,449 patients admitted with COVID-19 reported that 1993(36.6%) patients developed AKI during their hospitalization, of which 14.3% patients required renal replacement therapy(RRT).[79] Other clinical and laboratory manifestations include proteinuria, hematuria, electrolyte abnormalities such as hyperkalemia, hyponatremia, acid-base balance disturbance such as metabolic acidosis.[80][49][31]
- Cardiac manifestations: Myocardial injury manifesting as myocardial ischemia/infarction (MI) and myocarditis are well-recognized cardiac manifestations in patients with COVID-19. Other common cardiac manifestations include ACS, arrhythmias, cardiomyopathy, and cardiogenic shock. A single-center retrospective study analysis of 187 patients with confirmed COVID-19 reported that 27.8% of patients exhibited myocardial injury indicated by elevated troponin levels. The study also noted that patients with elevated troponin levels had more frequent malignant arrhythmias and a high mechanical ventilation rate than patients with normal troponin levels.[80] A meta-analysis study of 198 published studies involving 159, 698 COVID-19 patients reported that acute myocardial injury and high burden of pre-existing cardiovascular disease was significantly associated with higher mortality and ICU admission.[81]
- Hematologic manifestations: Lymphopenia is a common laboratory abnormality in the vast majority of patients with COVID-19. Other laboratory abnormalities include thrombocytopenia, leukopenia, elevated ESR levels, C-reactive protein (CRP) lactate dehydrogenase (LDH), and leukocytosis. As previously discussed, COVID-19 is also associated with a hypercoagulable, evidenced by the high prevalence of venous and thromboembolic events such as PE, DVT, MI, ischemic strokes, and arterial thromboses that also occurred in patients despite being maintained on prophylactic or even therapeutic systemic anticoagulation. Notably, COVID-19 is associated with markedly elevated D-dimer, fibrinogen levels, prolonged prothrombin time (PT), and partial thromboplastin time(aPTT) in patients at risk of developing arterial and venous thrombosis.[49][80][49] Clinical trials are required to determine the benefit of therapeutic anticoagulation in patients with COVID-19, especially at what stage of the illness.
- Gastrointestinal manifestations: GI symptoms such as diarrhea, nausea and/or vomiting, anorexia, and abdominal pain are seen in up to 1 in 5 patients with COVID-19 infection based on the results of a meta-analysis study by Tariq et al. that analyzed 78 studies involving 12, 797 patients. The weighted pool prevalence of diarrhea was 12.4% (95% CI, 8.2% to 17.1%), nausea and/or vomiting was 9% (95% CI, 5.5% to 12.9%), loss of appetite was 22.3% (95% CI, 11.2% to 34.6%) and abdominal pain was 6.2% (95% CI, 2.6% to 10.3%). The study also reported that the mortality rate among patients with GI symptoms was similar to the overall mortality rate.[82] Cases of acute mesenteric ischemia and portal vein thrombosis have also been described.[83]
- Hepatobiliary manifestations: Elevation in liver function tests manifesting as an acute increase in aspartate transaminase(AST) and alanine transaminase(ALT) are frequently noted in 14% to 53% of patients with COVID-19 infection.[84] Hepatic dysfunction occurs more frequently in patients with severe COVID-19 illness.
- Endocrinologic manifestations: Patients with underlying endocrinologic disorders such as diabetes mellitus who contract this virus are at increased risk of developing severe illness. Clinical manifestations such as abnormal blood glucose levels, euglycemic ketosis, and diabetic ketoacidosis have been noted in patients hospitalized with COVID-19.[80]
- Neurologic manifestations: Besides anosmia and ageusia, other neurological findings include headache, stroke, impairment of consciousness, seizure disorder, and toxic metabolic encephalopathy. Five patients with COVID-19 developed Guillain-Barré syndrome (GBS) based on a case series report from Northern Italy.[85][59]
- Cutaneous manifestations: Acral lesions resembling pseudo chilblains (40.4%) were the most common cutaneous manifestations noted in patients with COVID-19 based on the results of a meta-analysis study which included 34 published studies describing 996 patients with COVID-19. Other cutaneous manifestations described erythematous maculopapular rash (21.3%), vesicular rashes (13%), and urticarial rashes (10.9%). Notably, the appearance of a specific type was rash appeared to be dependent on the patient\’s age. Other uncommon rashes described were vascular rashes (4%) resembling livedo or purpura, especially in elderly patients, and erythema multiforme-like eruptions (3.7%), mostly in children.[86]
Evaluation
A detailed clinical history regarding the onset and duration of symptoms, travel history, exposure to people with COVID-19 infection, underlying preexisting medical conditions, and drug history should be elicited by treating providers. Patients with typical clinical signs suspicious of COVID-19 such as fever, cough, sore throat, loss of taste or smell, malaise, and myalgias should be promptly tested for SARS-CoV-2. Besides symptomatic patients, patients with atypical symptoms of COVID-19 or anyone with known high-risk exposure to SARS-CoV-2 should be tested for SARS-CoV-2 infection even in the absence of symptoms.
Diagnostic Testing Testing In COVID-19
Molecular Testing
-
The standard diagnostic mode of testing is testing a nasopharyngeal swab for SARS-CoV-2 nucleic acid using a real-time PCR assay. Commercial PCR assays have been validated by the US Food and Drug Administration (FDA) with emergency use authorizations (EUAs) for the qualitative detection of nucleic acid from SARS-CoV-2 from specimens obtained from nasopharyngeal swabs as well as other sites such as oropharyngeal, anterior/mid-turbinate nasal swabs, nasopharyngeal aspirates, bronchoalveolar lavage (BAL) and saliva. The collection of BAL samples should only be performed in mechanically ventilated patients as lower respiratory tract samples seem to remain positive for a more extended period.
- The sensitivity of PCR testing is dependent on multiple factors that include the adequacy of the specimen, technical specimen collection, time from exposure, and specimen source.[87] However, the specificity of most commercial FDA-approved SARS-CoV-2 PCR assays is nearly 100%, provided that there is no cross-contamination during specimen processing.
- SARS-CoV-2 antigen tests are less sensitive but have a faster turnaround time compared to molecular PCR testing.[88] Comprehensive testing for other respiratory viral pathogens should be considered for appropriate patients as well.
Serology Testing
-
An antibody test can evaluate for the presence of antibodies that occurs as a result of infection. Antibody tests play an important role in broad-based surveillance of COVID-19, and many commercial manufactured antibody testing kits are available to evaluate the presence of antibodies against SARS-CoV-2 are available.
-
Despite the numerous antibody tests designed to date, serologic testing has limitations in specificity and sensitivity, and results from different tests vary. However, an antibody test with a specificity higher than 99% and a sensitivity of 96% has been developed by the CDC, which can identify past SARS-CoV-2 infection.
-
Antibody testing may be instrumental in broad-based surveillance of COVID-19 and evaluate the immunity conferred from infection or vaccination. There is currently ongoing research to determine quantitative and qualitative aspects of antibodies regarding protection from future SARS-CoV-2 infection and the duration of the protection.
Other Laboratory Assessment
-
Complete blood count (CBC), a comprehensive metabolic panel (CMP) that includes testing for renal and liver function, and a coagulation panel should be performed in all hospitalized patients.
-
Additional tests such as testing for inflammatory markers such as ESR, C-reactive protein (CRP), ferritin, lactate dehydrogenase, D-dimer, and procalcitonin can be considered in hospitalized patients. However, their prognostic significance in COVID-19 is not clear.
Imaging Modalities
Considering this viral illness commonly manifests itself as pneumonia, radiological imaging has a fundamental role in the diagnostic process, management, and follow-up. Imaging studies may include chest x-ray, lung ultrasound, or chest computed tomography (CT). There are no guidelines available regarding the timing and choice of pulmonary imaging studies in patients with COVID-19, and the type of imaging should be considered based on clinical evaluation.
Chest X-ray
-
Standard radiographic examination (X-ray) of the chest has a low sensitivity in identifying early lung changes; it can be completely normal in the initial stages of the disease.
-
In the more advanced stages of infection, the chest X-ray examination commonly shows bilateral multifocal alveolar opacities, which tend to confluence up to the complete opacity of the lung. Pleural effusion can also be demonstrated.
Chest Computed Tomography (CT)
-
The American College of Radiology recommends against Chest CT\’s routine use as an initial imaging study or screening.
-
Given its high sensitivity, chest computed tomography (CT), particularly high-resolution CT (HRCT), is the diagnostic method of choice in evaluating COVID-19 pneumonia, particularly when associated with disease progression.
-
Several non-specific findings and radiologic patterns can be found on Chest CT. Most of these findings may also be observed in other lung infections, such as Influenza A (H1N1), CMV, SARS, MERS, streptococcus, and Chlamydia, Mycoplasma.
-
The most common CT findings in COVID-19 are multifocal bilateral \”ground or ground glass\” (GG) areas associated with consolidation areas with patchy distribution, mainly peripheral/subpleural, and greater involvement of the posterior regions lower lobes. The \”crazy paving\” pattern can also be observed.
-
This latter finding is characterized by GG areas with superimposed interlobular septal thickening and intralobular septal thickening. It is a non-specific finding that can be detected in different conditions.
-
Other notable findings include the \”reversed halo sign,\” a focal area of GG delimited by a peripheral ring with consolidation, and the findings of cavitations, calcifications, lymphadenopathies, and pleural effusion.
Lung Ultrasound
Ultrasonographic examination of the lung allows evaluating the progression of the disease, from a focal interstitial pattern up to a \”white lung\” with evidence of subpleural consolidations. Considering its noninvasive nature and zero risks of radiation, it is a useful diagnostic modality for patient follow-up and assists in determining the setting of mechanical ventilation and prone positioning. The main sonographic features are:
-
Pleural lines: appear often thickened, irregular, and discontinuous until it almost seems erratic; subpleural lesions can be seen as small patchy consolidations or nodules.
-
B lines: They are often motionless, coalescent, and cascade and can flow up to the square of \”white lung.\”
-
Thickenings: They are most evident in the posterior and bilateral fields, especially in the lower fields; the dynamic air bronchogram within the consolidation is a manifestation of disease evolution.
-
Perilesional pleural effusion
In summary, during the course of the illness, it is possible to identify the first phase with focal areas of fixed B lines followed by a phase of numerical increase of the lines B up to the white lung with small subpleural thickening, which progresses further until there is evidence of posterior consolidations.
Treatment / Management
Initially, early in the pandemic, the understanding of COVID-19 and its therapeutic management was limited, creating an urgency to mitigate this new viral illness with experimental therapies and drug repurposing. Since then, due to the intense efforts of clinical researchers globally, significant progress has been made, which has led to a better understanding of not only COVID-19 and its management but also has resulted in the development of novel therapeutics and vaccine development at an unprecedented speed.
Pharmacologic Therapies In The Management Of Adults With COVID-19
Currently, a variety of therapeutic options are available that include antiviral drugs (e.g., remdesivir), anti-SARS-CoV-2 monoclonal antibodies (e.g., bamlanivimab/etesevimab, casirivimab/imdevimab), anti-inflammatory drugs (e.g., dexamethasone), immunomodulators agents (e.g., baricitinib, tocilizumab) are available under FDA issued Emergency Use Authorization( EUA) or being evaluated in the management of COVID-19.[49]
The clinical utility of these treatments is specific and is based on the severity of illness or certain risk factors. The clinical course of the COVID-19 illness occurs in 2 phases, an early phase when SARS-CoV-2 replication is greatest before or soon after the onset of symptoms. Antiviral medications and antibody-based treatments are likely to be more effective during this stage of viral replication. The later phase of the illness is driven by a hyperinflammatory state induced by the release of cytokines and the coagulation system’s activation that causes a prothrombotic state. Anti-inflammatory drugs such as corticosteroids, immunomodulating therapies, or a combination of these therapies may help combat this hyperinflammatory state than antiviral therapies.[88] Below is a summary of the latest potential therapeutic options proposed, authorized, or approved for clinical use in the management of COVID-19.
Antiviral Therapies
- Remdesivir is a broad-spectrum antiviral agent that previously demonstrated antiviral activity against SARS-CoV-2 in vitro.[89] Based on results from three randomized, controlled clinical trials that showed that remdesivir was superior to placebo in shortening the time to recovery in adults who were hospitalized with mild-to-severe COVID-19, the U.S. Food and Drug Administration (FDA) approved remdesivir for clinical use in adults and pediatric patients (over age 12 years and weighing at least 40 kilograms or more) to treat hospitalized patients with COVID-19.[90][91][92] However, results from the WHO SOLIDARITY Trial conducted at 405 hospitals spanning across 40 countries involving 11,330 inpatients with COVID-19 who were randomized to receive remdesivir (2750) or no drug (4088) found that remdesivir had little or no effect on overall mortality, initiation of mechanical ventilation, and length of hospital stay.[93] There is no data available regarding the efficacy of remdesivir against the new SARS-CoV-2 variants; however, acquired resistance against mutant viruses is a potential concern and should be monitored.
- Hydroxychloroquine and chloroquine were proposed as antiviral treatments for COVID-19 initially during the pandemic. However, data from randomized control trials evaluating the use of hydroxychloroquine with or without azithromycin in hospitalized patients did not improve the clinical status or overall mortality compared to placebo.[94][93] Data from randomized control trials of hydroxychloroquine used as postexposure prophylaxis did not prevent SARS-CoV-2 infection or symptomatic COVID-19 illness.[95][96][96]
- Lopinavir/ritonavir is an FDA-approved combo therapy for the treatment of HIV and was proposed as antiviral therapy against COVID-19 during the early onset of the pandemic. Data from a randomized control trial that reported no benefit was observed with lopinavir-ritonavir treatment compared to standard of care in patients hospitalized with severe COVID-19.[97] Lopinavir/Ritonavir is currently not indicated for the treatment of COVID-19 in hospitalized and nonhospitalized patients.
- Ivermectin is an FDA-approved anti-parasitic drug used worldwide in the treatment of COVID-19 based on an in vitro study that showed inhibition of SARS-CoV-2 replication.[98] A single-center double-blind, randomized control trial involving 476 adult patients with mild COVID-19 illness was randomized to receive ivermectin 300 mcg/kg body weight for five days or placebo did not achieve significant improvement or resolution of symptoms.[99] Ivermectin is currently not indicated for the treatment of COVID-19 in hospitalized and nonhospitalized patients.
Anti-SARS-CoV-2 Neutralizing Antibody Products
Individuals recovering from COVID-19 develop neutralizing antibodies against SARS-CoV-2, and the duration of how long this immunity lasts is unclear. Nevertheless, their role as therapeutic agents in the management of COVID-19 is extensively being pursued in ongoing clinical trials.
- Convalescent Plasma therapy was evaluated during the SARS, MERS, and Ebola epidemics; however, it lacked randomized control trials to back its actual efficacy. The FDA approved convalescent plasma therapy under a EUA for patients with severe life-threatening COVID-19.[100][101] Although it appeared promising, data from multiple studies evaluating the use of convalescent plasma in life-threatening COVID-19 has generated mixed results. One retrospective study based on a U.S. national registry reported that among patients hospitalized with COVID-19, not on mechanical ventilation, there was a lower risk of death in patients who received a transfusion of convalescent plasma with higher anti-SARS-CoV-2 IgG antibody than patients who received a transfusion of convalescent plasma with low antibody levels. Data from three small randomized control trials showed no significant differences in clinical improvement or overall mortality in patients treated with convalescent plasma versus standard therapy.[102][103][104] An in vitro analysis of convalescent plasma obtained from individuals previously infected with the ancestral SARS-CoV-2 strains demonstrated significantly reduced neutralization against SARS-CoV-2 variant B.1.351/ 501Y.V2.[105] Another in vitro study reported B.1.351 variant exhibited markedly more resistance to neutralization by convalescent plasma obtained from individuals previously infected with the ancestral SARS-CoV-2 strains compared to the B.1.1.7 variant, which was not more resistant to neutralization.[106]
- REGN-COV2 (Casirivimab and Imdevimab): REGN-COV2 is an antibody cocktail containing two noncompeting IgG1 antibodies (casirivimab and imdevimab) that target the RBD on the SARS-CoV-2 spike protein that has been shown to decrease the viral load in vivo, preventing virus-induced pathological sequelae when administered prophylactically or therapeutically in non-human primates.[107] Results from an interim analysis of 275 patients from an ongoing double-blinded trial involving non hospitalized patients with COVID-19 who were randomized to receive placebo, 2.4 g of REGN-COV2 (casirivimab 1,200 mg and imdevimab 1,200 mg) or 8 g of REGN-COV2 COV2 (casirivimab 2,400 mg and imdevimab 2,400 mg) reported that the REGN-COV2 antibody cocktail reduced viral load compared to placebo. This interim analysis also established the safety profile of this cocktail antibody, similar to that of the placebo group.[108] Preliminary data from a Phase 3 trial of REGN-COV (casirivimab/imdevimab) revealed a 70% reduction in hospitalization or death in nonhospitalized patients with COVID-19. In vitro data is available regarding the effect of REGN-COV2 on the two new SARS-CoV-2 variants of concern (B.1.1.7; B.1.351 variants) that reveal retained activity.
- Bamlanivimab and Etesevimab (LY-CoV555 or LY3819253 and LY-CoV016 or LY3832479) are potent anti-spike neutralizing monoclonal antibodies. Bamlanivimab is a neutralizing monoclonal antibody derived from convalescent plasma obtained from a patient with COVID-19. Like REGN-COV2, it also targets the RBD of the spike protein of SARS-CoV-2 and has been shown to neutralize SARS-CoV-2 and reduce viral replication in non-human primates.[94][95] In vitro experiments revealed that etesevimab binds to a different epitope than bamlanivimab and neutralizes resistant variants with mutations in the epitope bound by bamlanivimab. In Phase 2 of the BLAZE-1 trial, bamlanivimab/etesevimab was associated with a significant reduction in SARS-CoV-2 viral load compared to placebo.[109] Data from the Phase 3 portion of BLAZE-1 is pending release, but preliminary information indicates that therapy reduced the risk of hospitalization and death by 87%. In vitro data is available regarding the effect of bamlanivimab/etesevimab on the new SARS-CoV-2 variants of concern (B.1.1.7; B.1.351) reveals retained activity.[110]
-
REGN-COV2 (casirivimab and imdevimab) and bamlanivimab/etesevimab were approved for clinical use by the FDA under two separate EUAs issued in November 2020 and February 2021, respectively, that allowed the use of these drugs only in nonhospitalized patients (aged ≥12 years and weighing ≥40 kg) with laboratory-confirmed SARS-CoV-2 infection and mild to moderate COVID-19 who are at high risk for progressing to severe disease and/or hospitalization.
Immunomodulatory Agents
- Corticosteroids: Severe COVID-19 is associated with inflammation-related lung injury driven by the release of cytokines characterized by an elevation in inflammatory markers. During the pandemic’s early course, glucocorticoids’ efficacy in patients with COVID-19 was not well described. The Randomized Evaluation of Covid-19 Therapy (RECOVERY) trial, which included hospitalized patients with clinically suspected or laboratory-confirmed SARS-CoV-2 who were randomly assigned to received dexamethasone (n=2104) or usual care (n=4321), showed that the use of dexamethasone resulted in lower 28-day mortality in patients who were on invasive mechanical ventilation or oxygen support but not in patients who were not receiving any respiratory support.[111] Based on this landmark trial results, dexamethasone is currently considered the standard of care either alone or in combination with remdesivir based on the severity of illness in hospitalized patients who require supplemental oxygen or non-invasive or invasive mechanical ventilation.
- Interferon-β-1a (IFN- β-1a): Interferons are cytokines that are essential in mounting an immune response to a viral infection, and SARS-CoV-2 suppresses its release in vitro.[112] However, previous experience with IFN- β-1a in acute respiratory distress syndrome (ARDS) has not benefited.[113] Results from a small randomized, double-blind, placebo-controlled trial showed the use of inhaled IFN- β-1a had greater odds of clinical improvement and recovery compared to placebo.[114] Another small randomized clinical trial showed that the clinical response using inhaled IFN- β-1a was not significantly different from the control group. The authors reported when used early, this agent resulted in a shorter length of hospitalization stay and decreased 28-day mortality rate. However, four patients who died in the treatment group before completing therapy were excluded, thus making the interpretation of these results difficult.[115] Currently, there is no data available regarding the efficacy of interferon β-1a on the three new SARS-CoV-2 variants (B.1.1.7; B.1.351 and P.1). Given the insufficient and small amount of data regarding this agent’s use and the relative potential for toxicity, this therapy is not recommended to treat COVID-19 infection.
- Interleukin (IL)-1 Antagonists: Anakinra is an interleukin-1 receptor antagonist that is FDA approved to treat rheumatoid arthritis. Its off-label use in severe COVID-19 was assessed in a small case-control study trial based on the rationale that the severe COVID-19 is driven by cytokine production, including interleukin (I.L.)-1β. This trial revealed that of the 52 patients who received anakinra and 44 patients who received standard of care, anakinra reduced the need for invasive mechanical ventilation and mortality in patients with severe COVID-19.[116] There is no data available regarding the efficacy of interleukin-1 receptor antagonists on the three new SARS-CoV-2 variants (B.1.1.7; B.1.351, and P.1). Given the insufficient data regarding this treatment based on case series only, this is not currently recommended to treat COVID-19 infection.
- Anti-IL-6 receptor Monoclonal Antibodies: Interleukin-6 (IL-6) is a proinflammatory cytokine that is considered the key driver of the hyperinflammatory state associated with COVID-19. Targeting this cytokine with an IL-6 receptor inhibitor could slow down the process of inflammation based on case reports that showed favorable outcomes in patients with severe COVID-19.[48][117][118] The FDA approved three different types of IL-6 receptor inhibitors for various rheumatological conditions (Tocilizumab, Sarilumab) and a rare disorder called Castleman’s syndrome (Siltuximab).
- Tocilizumab is an anti-interleukin-6 receptor alpha receptor monoclonal antibody that has been indicated for various rheumatological diseases. The data regarding the use of this agent is mixed. A randomized control trial involving 438 hospitalized patients with severe COVID-19 pneumonia, among which 294 were randomized to receive tocilizumab and 144 to placebo, showed that tocilizumab did not translate into a significant improvement in clinical status or lower the 28-day mortality compared to placebo.[119] Results from another randomized, double-blind placebo-controlled trial involving patients with confirmed severe COVID-19 that involved 243 patients randomized to receive tocilizumab or placebo showed that the use of tocilizumab was not effective in preventing intubation or death rate.[120] The REMAP-CAP and RECOVERY trials (not yet published), two large randomized controlled trials, showed a mortality benefit in patients exhibiting rapid respiratory decompensation.[121]
- Sarilumab and Siltuximab are IL-6 receptor antagonists that may potentially have a similar effect on the hyperinflammatory state associated with COVID-19 as tocilizumab. Currently, there no known published clinical trials supporting the use of siltuximab in severe COVID-19. Conversely, a 60-day randomized, double-blind placebo control multinational phase 3 trial that evaluated the clinical efficacy, mortality, and safety of sarilumab in 431 patients did not show any significant improvement in clinical status or mortality rate.[122] Another randomized, double-blind placebo-controlled study on sarilumab’s clinical efficacy and safety in adult patients hospitalized with COVID-19 is currently ongoing (NCT04315298).
- Baricitinib is an oral selective inhibitor of Janus kinase (JAK) 1 and JAK 2 currently indicated for moderate to severely active rheumatoid arthritis patients. Baricitinib was considered a potential treatment for COVID-19 based on its inhibitory effect on SARS-CoV-2 endocytosis in vitro and on the intracellular signaling pathway of cytokines that cause the late-onset hyperinflammatory state that results in severe illness.[123][124][123] This dual inhibitory effect makes it a promising therapeutic drug against all stages of COVID-19. A multicenter observational, retrospective study of 113 hospitalized patients with COVID-19 pneumonia who received baricitinib combined with lopinavir/ritonavir (baricitinib arm, n=113) or hydroxychloroquine and lopinavir/ritonavir (control arm, n=78) reported significant improvement in clinical symptoms and 2-week mortality rate in the baricitinib arm compared with the control arm. Results from the ACTT-2 trial, a double-blind, randomized placebo-controlled trial evaluating baricitinib plus remdesivir in hospitalized adult patients with COVID-19, reported that the combination therapy of baricitinib plus remdesivir was superior to remdesivir therapy alone in not only reducing recovery time but also accelerating clinical improvement in hospitalized patients with COVID-19, particularly who were receiving high flow oxygen supplementation or noninvasive ventilation.[125] Baricitinib, in combination with remdesivir, has been approved for clinical use in hospitalized patients with COVID-19 under a EUA issued by the FDA. The efficacy of baricitinib alone or in combination with remdesivir has not been evaluated in the SARS-CoV-2 variants, and there is limited data on the use of baricitinib with dexamethasone.
- Ruxolitinib is another oral selective inhibitor of JAK 1 and 2 that is indicated for myeloproliferative disorders, polycythemia vera, and steroid-resistant GVHD. Similar to baricitinib, it has been hypothesized to have an inhibitory effect on cytokines’ intracellular signaling pathway, making it a potential treatment against COVID-19. Results from a small prospective multicenter randomized controlled phase 2 trial evaluating the efficacy and safety of ruxolitinib reported no statistical difference than the standard of care. However, most of the patients demonstrated significant chest C.T. improvement and faster recovery from lymphopenia.[126] A large randomized, double-blind, placebo-controlled multicenter trial (NCT04362137) is ongoing to assess ruxolitinib’s efficacy and safety in patients with severe COVID-19.
- Bruton’s tyrosine kinase inhibitors such as acalabrutinib, ibrutinib, rilzabrutinib are tyrosine kinase inhibitors that regulate macrophage signaling and activation currently FDA approved for some hematologic malignancies. It is proposed that macrophage activation occurs during the hyperinflammatory immune response seen in severe COVID-19. Results from a small off-label study of 19 hospitalized patients with severe COVID-19 who received acalabrutinib highlighted the potential clinical benefit of BTK inhibition.[127] Clinical trials are in progress to validate the actual efficacy of these drugs in severe COVID-19 illness.
Oxygenation And Ventilation Management In COVID-19
Conventional Oxygen Therapy
COVID-19 patients with associated respiratory insufficiency should be monitored closely with continuous pulse oximetry. Supplemental oxygen supplementation via nasal cannula or Venturi mask must be administered to maintain oxygen saturation (SpO2) between 92 to 96% (< 88-90% if COPD). If there is improvement in clinical and oxygen saturation, supplemental oxygen should be continued with periodic reassessment. If there is no clinical improvement or worsening of symptoms and/or oxygen saturation, non-invasive treatments such as High-Flow Nasal Cannula (HFNC) or Noninvasive Positive Pressure Ventilation(NIPPV) are recommended.
Management of Acute Hypoxemic Respiratory Failure in COVID-19
Acute hypoxemic respiratory failure is the most common complication in adult patients with COVID-19, and conventional oxygen therapy is not helpful to address the oxygen demand in these patients. These patients should be managed with enhanced respiratory support modalities such as high-flow nasal cannula (HFNC), noninvasive positive pressure ventilation (NIPPV), endotracheal intubation, and invasive mechanical ventilation (IMV) or extracorporeal membrane oxygenation (ECMO)
High-Flow Nasal Cannula (HFNC) and Noninvasive Positive Pressure Ventilation (NIPPV)
HFNC and NIPPV are noninvasive enhanced respiratory support modalities available in managing COVID-19-associated acute hypoxemic respiratory failure and are instrumental in avoiding invasive mechanical ventilation in carefully selected patients. A meta-analysis study evaluating the effectiveness of HFNC compared to conventional oxygen therapy and NIPPV before mechanical ventilation reported that HFNC, when used before mechanical ventilation, could improve the prognosis of patients compared to conventional oxygen therapy and NIPPV.[128] Use of HFNC or NIPPV is associated with decreased dispersion of exhaled air especially when used with a good interface fitting, thus creating a low risk of nosocomial transmission of the infection.[129] However, these treatment modalities are associated with a greater risk of aerosolization and should be used in negative pressure rooms.
Noninvasive Positive-pressure Ventilation (NIPPV)
-
NIPPV (bilevel positive airway pressure [BiPAP]/continuous positive airway pressure [CPAP]) is instrumental in the management of COVID-19-associated acute hypoxemic respiratory failure and may help avoid invasive mechanical ventilation in carefully selected patients.
- NIPPV should be restricted to hospitalized patients with COVID-19 who develop respiratory insufficiency due to COPD, cardiogenic pulmonary edema, or have underlying obstructive sleep apnea (OSA) rather than ARDS.[130]
-
A helmet is preferred for minimizing the risk of aerosolization. In NIPPV with face masks (full-face or oronasal), the use of masks integrated with an expiratory valve fitted with an antimicrobial filter is recommended.
-
Results from the HENIVOT trial, an Italian open-label multicenter randomized clinical trial, reported that there was no significant difference in the number of days free of respiratory support with the utilization of helmet noninvasive ventilation treatment compared to high flow nasal oxygen in COVID-19 patients hospitalized with moderate to severe degree of hypoxemia.
Endotracheal Intubation and Lung Protective Invasive Mechanical Ventilation
- Impending respiratory failure should be recognized as early as possible, and a skilled operator must promptly perform endotracheal intubation to maximize first-pass success.[131]
- Clinicians and other healthcare staff must wear appropriate PPE that includes gowns, gloves, N95 masks, and eye protection when performing endotracheal intubation and manual ventilation before intubation, physical proning of the patient, or providing critical patient care such as upper airway suctioning, disconnecting the patient from the ventilator.[131]
-
Preoxygenation (100% O2 for 5 minutes) should be performed via HFNC.
-
Invasive mechanical ventilation in COVID-19 associated acute hypoxemic respiratory failure and ARDS should be with lower tidal volumes (V.T.) (4 to 8 ml/kg predicted body weight, PBW) and lower inspiratory pressures reaching a plateau pressure (Pplat) < 30 cm of H2O.
-
Positive end-expiratory pressure (PEEP) must be as high as possible to maintain the driving pressure (Pplat-PEEP) as low as possible (< 14 cmH2O).
-
Use of neuromuscular blocking agents (NMBA) should be used as needed to facilitate lung-protective ventilation.
-
In patients with refractory hypoxemia (PaO2:FiO2 of 12 to 16 hours per day and the use of a conservative fluid management strategy for ARDS patients without tissue hypoperfusion are strongly emphasized.
-
The National Institutes of Health (NIH) Covid-19 Treatment Guidelines Panel recommends against inhaled pulmonary vasodilators such as nitric oxide.
-
Lung-protective ventilation can also reduce the risk of new or worsening AKI by preventing ventilator-induced hemodynamic effects.
-
ECMO should be considered in carefully selected patients with refractory hypoxemia despite lung-protective ventilation and patients who fail to respond to prone position ventilation.
Management Of COVID-19 Based On The Severity of Illness
-
Mild Illness
-
-
Based on the NIH guidelines, individuals with mild illness is manageable in the ambulatory setting with supportive care and isolation.
-
Laboratory and radiographic evaluation are routinely not indicated.
-
Elderly patients and those with pre-existing conditions should be monitored closely until clinical recovery is achieved.
-
SARS-CoV-2 neutralizing antibodies such as REGN-COV2 (casirivimab and imdevimab) or bamlanivimab/etesevimab can be considered for outpatients who are at risk of disease progression with a low threshold to consider hospitalization for closer monitoring.
-
The National Institutes of Health (NIH) Covid-19 Treatment Guidelines Panel recommends against dexamethasone in mild illness.
-
-
Moderate Illness
-
-
Patients with moderate COVID-19 illness should be hospitalized for close monitoring.
-
Clinicians and healthcare staff should don appropriate personal protective equipment (PPE) while interacting or taking care of the patient.
- All hospitalized patients should receive supportive care with isotonic fluid resuscitation if volume-depleted, and supplemental oxygen therapy must be initiated if SpO2 and be maintained no higher than 96%.[132]
-
Empirical antibacterial therapy should be started only if there is a suspicion of bacterial infection and should be discontinued as early as possible if not indicated.
-
Patients with COVID-19 are at risk of developing venous and thromboembolic events and should be maintained on thromboembolic prophylaxis with appropriate anticoagulation.
-
Remdesivir and dexamethasone can be considered for patients who are hospitalized and require supplemental oxygen.
-
The National Institutes of Health (NIH) Covid-19 treatment guidelines panel recommends the use of either remdesivir alone or dexamethasone plus remdesivir or dexamethasone alone if combination therapy (remdesivir and dexamethasone) is not available in hospitalized patients who require supplemental oxygen but are not receiving HFNC or NIPPV or IMV or ECMO.
-
-
-
Patients with severe/critical COVID-19 illness require hospitalization.
-
Considering that patients with severe COVID-19 are at increased risk of prolonged critical illness and death, discussions regarding care goals, reviewing advanced directives, and identifying surrogate medical decision-makers must be made.
-
All patients should be maintained on prophylactic anticoagulation, considering COVID-19 is associated with a prothrombotic state.
- Clinicians and other healthcare staff must wear appropriate PPE that include gowns, gloves, N95 masks, and eye protection when performing aerosol-generating procedures on patients with COVID-19 in the ICU, such as endotracheal intubation, bronchoscopy, tracheostomy, manual ventilation before intubation, physical proning of the patient or providing critical patient care such as nebulization, upper airway suctioning, disconnecting the patient from the ventilator, and noninvasive positive pressure ventilation that may potentially lead to the aerosol generation.[132]
-
Renal replacement therapy should be considered in renal failure when indicated.
-
HFNC or NIPPV can be considered in patients who do not require intubation.
-
Having awake patients self-prone while receiving HFNC can improve oxygenation if endotracheal intubation is not indicated. However, the efficacy of performing this maneuver on awake patients is not clear and more data from clinical trials is needed.
-
The National Institutes of Health (NIH) Covid-19 Treatment Guidelines Panel strongly recommends using dexamethasone in hospitalized patients who require oxygen via noninvasive or invasive ventilation. Combination therapy with dexamethasone plus remdesivir can also be an option. If corticosteroids cannot be used, baricitinib plus remdesivir may be used.
-
The National Institutes of Health (NIH) Covid-19 Treatment Guidelines Panel also recommends tocilizumab (as a single intravenous dose) in recently hospitalized patients who are exhibiting rapid respiratory decompensation due to COVID-19.
-
Impending respiratory failure should be recognized as early as possible, and endotracheal intubation with IMV must be initiated as described earlier.
-
Vasopressors should be started to maintain mean arterial pressure (MAP) between 60 mmHg and 65 mmHg. Norepinephrine is the preferred initial vasopressor.
-
Empiric antibacterial therapy should be considered if there is a concern for a secondary bacterial infection. Antibiotic use must be reassessed daily for de-escalation, and the duration of the treatment requires evaluation for appropriateness based on the diagnosis.
- Management of COVID-19 patients with ARDS should be similar to classical ARDS management from other causes, including prone positioning as per The Surviving Sepsis Campaign guidelines for managing COVID-19.[132]
-
ECMO should be considered in patients with refractory respiratory failure as previously described
-
Prevention Of COVID-19
Besides the importance of imposing public health and infection control measures to prevent or decrease the transmission of SARS-CoV-2, the most crucial step to contain this global pandemic is by vaccination to prevent SARS-CoV-2 infection in communities across the world. Extraordinary efforts by clinical researchers worldwide during this pandemic have resulted in the development of novel vaccines against SARS-CoV-2 at an unprecedented speed to contain this viral illness that has devastated communities worldwide. Vaccination triggers the immune system leading to the production of neutralizing antibodies against SARS-CoV-2.
BNT162b2 vaccine: Results of an ongoing multinational, placebo-controlled, observer-blinded, pivotal efficacy trial reported that individuals 16 years of age or older receiving two-dose regimen the trial vaccine BNT162b2 (mRNA-based, BioNTech/Pfizer) when given 21 days apart conferred 95% protection against COVID-19 with a safety profile similar to other viral vaccines.[133] Based on the results of this vaccine efficacy trial, the FDA issued a EUA on December 11, 2020, granting the use of the BNT162b2 vaccine to prevent COVID-19.
mRNA-1273 vaccine: Results from another multicenter, Phase 3, randomized, observer-blinded, placebo-controlled trial demonstrated that individuals who were randomized to receive two doses of mRNA-1273 (mRNA based, Moderna) vaccine given 28 days apart showed 94.1% efficacy at preventing COVID-19 illness and no safety concerns were noted besides transient local and systemic reactions.[134] Based on the results of this vaccine efficacy trial, the FDA issued a EUA on December 18, 2020, granting the use of the mRNA-1273 vaccine to prevent COVID-19.
Ad26.COV2.S vaccine: A third vaccine Ad26.COV2.S vaccine for the prevention of COVID-19 received EUA by the FDA on February 27, 2021, based on a multicenter, placebo control, phase trial showed that a single dose of Ad26.COV2.S vaccine conferred 73% efficacy in the U.S. in preventing COVID-19 (data not yet published)
ChAdOx1 nCoV-19:Interim analysis of an ongoing multicenter randomized control trial demonstrated clinical efficacy against symptomatic COVID-19 and had an acceptable safety profile.[135] The ChAdOx1 nCoV-19 vaccine has been approved or granted emergency use authorization to prevent COVID-19 in many countries across the world but has not yet received a EUA or approval from the FDA for use in the U.S.
In addition to the vaccines mentioned above, as many as seven other vaccines, including protein-based and inactivated vaccines, have been developed indigenously in India, Russia, and China and have been approved or granted emergency use authorization to prevent COVID-19 in many countries around the world.
Differential Diagnosis
The symptoms of the early stages of the disease are nonspecific. Differential diagnosis should include the possibility of a wide range of infectious and non-infectious (e.g., vasculitis, dermatomyositis) respiratory disorders.
For suspected cases, rapid antigen detection and other investigations should be adopted for evaluating common respiratory pathogens and non-infectious conditions.
Pertinent Studies and Ongoing Trials
Efficacy Of Available COVID-19 Vaccines In Prevention Against SARS-CoV-2 Variants Of Concern
The three novel vaccines, BNT162b2 vaccine, mRNA-1273 vaccine, and ChAdOx1 nCoV-19, were developed to target the SARS-CoV-2 spike protein main site where these variants have developed mutations, raising concerns regarding the efficacy of these vaccines against the new variants.
- BNT162b2 vaccine: The efficacy of the BNT162b2 vaccine against the three SARS-CoV-2 variants is unknown. In vitro analysis of 20 serum samples obtained from 15 participants from the BNT162b2 clinical efficacy trial efficiently neutralized all SARS-CoV-2 variants. Neutralization of B.1.1.7 variant and P.1 was roughly equivalent. The neutralization of B.1.351 was vigorous but lower than the ancestral SARS-CoV-2 strain.[136][133][136] Clinical trials of the BNT162b2 vaccine against these three new SARS-CoV-2 variants are ongoing and are awaited.
- mRNA-1273 vaccine: The efficacy of the mRNA-1273 vaccine against the SARS-CoV-2 variants is unknown. In vitro analysis of serum samples obtained from participants of the mRNA-1273 vaccine, clinical efficacy trial demonstrated that the mutations affecting the RBD of the B.1.1.7 variant had no significant effect on neutralization by serum obtained from participants who received the mRNA-1273 vaccine. Conversely, the analysis also showed a decrease in titers of neutralizing antibodies against the B.1.1.7+E484K variant, B.1.351 variant, P.1 variant, and the B.1.427/B.1.429 variants. The reduction in neutralizing titers was significantly lower in the B.1.351 variant.[137]
- Ad26.COV2.S vaccine: A single dose of this vaccine offers protection against COVID-19 consistently across many countries, including Brazil with a predominant percentage of strains from the P.2 lineage and across South Africa with a predominant percentage of strains from the B.1.135 lineage (data not yet reported). It is important to note that the vaccine\’s efficacy in the US was higher by a factor of 1.3 compared to South Africa (72% versus 57%).[138]
- ChAdOx1 nCoV-19 vaccine: A two-dose regimen of the ChAdOx1 nCoV-19 vaccine did not confer protection against mild to moderate COVID-19 SARS-CoV-2 B.1.351 variant based on results from a multicenter, double-blind, randomized control trial 33725432. Results of another randomized control trial regarding the ChAdOx1 nCoV-19 vaccine showed that in vitro neutralization activity against the B.1.1.7 variant was reduced compared with a non-B.1.1.7 variant and the clinical efficacy of the vaccine was 70.4 % for B.1.1.7 showed compared to 81.5 % efficacy noted in non-B.1.1.7 variants.[139]
Prognosis
The prognosis of COVID-19 is largely dependent on various factors that include the patient\’s age, the severity of illness at presentation, pre-existing conditions, how quickly treatment can be implemented, and response to treatment. As previously described, the WHO’s current estimate of the global case fatality rate for COVID-19 is 2.2%. However, the case fatality rate is affected by factors such as age, underlying pre-existing conditions, and severity of illness. Results from a European multicenter prospective cohort study that included 4000 critically ill patients with COVID-19 reported a 90-day mortality of 31%, with higher mortality noted in elderly, diabetic, obese, and severe ARDS patients[140]
Complications
COVID-19 can be regarded as a systemic viral illness based on its involvement in multiple major organ systems.
-
Patients with advanced age and comorbid conditions such as obesity, diabetes mellitus, chronic lung disease, cardiovascular disease, chronic kidney disease, chronic liver disease, and neoplastic conditions are at risk of developing severe COVID-19 and its associated complications. The most common complication of severe COVID-19 illness is progressive or sudden clinical deterioration leading to acute respiratory failure and ARDS and/or multiorgan failure leading to death.
- Patients with COVID-19 illness are also at increased risk of developing prothrombotic complications such as PE, DVT, MI, ischemic strokes, and arterial thrombosis.[49]
-
Cardiovascular system involvement results in malignant arrhythmias, cardiomyopathy, and cardiogenic shock.
- GI complications such as bowel ischemia, transaminitis, gastrointestinal bleeding, pancreatitis, Ogilvie syndrome, mesenteric ischemia, and severe ileus are often noted in critically ill patients with COVID-19 [141]
- Acute renal failure is the most common extrapulmonary manifestation of COVID-19 and is associated with an increased risk of mortality.[78]
- A meta-analysis study of 14 studies evaluating the prevalence of disseminated intravascular coagulation (DIC) in hospitalized patients with COVID-19 reported that DIC was observed in 3% (95%: 1%-5%, P [142]
- More recent data have emerged regarding prolonged symptoms in patients who have recovered from COVID-19 infection, termed \”post-acute COVID-19 syndrome.\” A large cohort study of 1773 patients performed 6 months after hospitalization with COVID-19 revealed that most exhibited at least one persistent symptom: fatigue, muscle weakness, sleep difficulties, or anxiety. Patients with severe illness also had an increased risk of chronic lung issues.[143]
- A retrospective cohort study that included 236,379 patients reported substantial neurological (intracranial hemorrhage, ischemic stroke) and psychiatric morbidity (anxiety disorder, psychotic disorder) 6 months after being diagnosed with COVID-19.[144]
Deterrence and Patient Education
-
Patients and families must be educated and encouraged to adhere to social distancing guidelines, use of facemasks and travel guidelines as per CDC guidelines, and social distancing state and local authorities\’ social distancing protocols.
-
Patients must be educated about frequent handwashing for a minimum of 20 seconds with soap and water when they come in contact with contaminated surfaces.
-
Patients should be educated and encouraged to seeking emergency care when necessary.
-
Patients should be educated and given an option for telehealth services in place of office visits if applicable.
-
High-risk patients should be encouraged to seek treatment early and be educated on new treatment options such as monoclonal antibodies.
-
Patients require education regarding the efficacy of the available vaccines and the benefits of the vaccination.
Enhancing Healthcare Team Outcomes
-
COVID-19 has had a straining effect on many healthcare systems across the world. Three vaccines have been authorized for use in the US by the FDA under an Emergency Use Authorization (EUA), and other approvals have been issued worldwide.
-
Until most of the world’s population gets vaccinated against this illness, COVID-19 will continue to remain a threat to global public health with the emergence of potentially treatment-resistant variants.
-
Prevention and management of this highly transmissible respiratory viral illness require a holistic and interprofessional approach that includes physicians\’ expertise across specialties, nurses, pharmacists, public health experts, and governmental authorities. There should be closed-loop communication between the clinical providers, pharmacists, and nursing staff while managing patients with COVID-19.
-
Clinical providers managing COVID-19 patients on the frontlines should keep themselves periodically updated with the latest clinical guidelines about diagnostic and therapeutic options available in the management of COVID-19 especially considering the emergence of new SARS-CoV-2 variants, which could have a huge impact on morbidity and mortality.
-
Clinicians should maintain a high index of suspicion in patients from a high risk of exposure area or recent travel to a high exposure area who present with extrapulmonary manifestations in the absence of pulmonary symptoms. These patients should be appropriately triaged and tested for SARS-CoV-2.
-
Resources for contact tracing and testing must be enhanced to limit the spread of this virus. Patients must be educated and encouraged to adhere to social distancing guidelines, travel guidelines, and the use of facemasks as per CDC guidelines and COVID-19 protocols of state and local authorities.
-
Clinical pharmacists must also keep themselves updated about the emergence of novel therapeutics that have been approved or granted emergency use authorization in the management of COVID-19.
-
There must be a strong focus to educate the public about the importance of receiving the vaccination against COVID-19, and consideration must be made to establish mass vaccination sites.
-
Continued viral surveillance of new variants is crucial at regular intervals with viral genomic sequencing given the possibility that more highly transmissible, more virulent variants and treatment-resistant variants could emerge that can have a more catastrophic effect on global health in addition to the current scenario.
-
Such a multi-pronged approach enhances improved patient care and outcomes. It also reduces the burden of hospitalizations that could potentially lead to the exhaustion of healthcare resources.
-
Hospitals and communities should have in place a plan to triage moderate and high-risk patients for additional therapy, such as monoclonal antibodies, on an outpatient basis.
-
Such interprofessional team measures could immensely change the dynamic of healthcare infrastructure and go a long way in eradicating or eliminating this virus and limiting its devastating effect on socioeconomic and healthcare situations across the entire world.
The interprofessional healthcare team will include all public health authorities, clinicians, specialists, mid-level practitioners, nursing staff, pharmacists, and even the patients and potential patients of this illness, all working collaboratively and openly sharing information bring about positive outcomes both for individual patients as well as society as a whole. [Level 5]
Continuing Education / Review Questions
Figure
Covid 19, Corona Replication. Contributed by Rohan Bir Singh, MD
Figure
Clinical Presentation of Patients with CoVID-19. Contributed by Rohan Bir Singh, MD; Made with Biorender.com
Figure
SARS- CoV 2 Structure. Contributed by Rohan Bir Singh, MD; Made with Biorender.com
Figure
Transmission Cycle of SARS CoV 2. Contributed by Rohan Bir Singh, MD; Made with Biorender.com
Figure
Single-stranded RNA genome of SARS-CoV2. Contributed by Rohan Bir Singh, MD; Made with Biorender.com
References
1.Chan JF, To KK, Tse H, Jin DY, Yuen KY. Interspecies transmission and emergence of novel viruses: lessons from bats and birds. Trends Microbiol. 2013 Oct;21(10):544-55. [PMC free article: PMC7126491] [PubMed: 23770275]2.Lei J, Kusov Y, Hilgenfeld R. Nsp3 of coronaviruses: Structures and functions of a large multi-domain protein. Antiviral Res. 2018 Jan;149:58-74. [PMC free article: PMC7113668] [PubMed: 29128390]3.Chan JF, Kok KH, Zhu Z, Chu H, To KK, Yuan S, Yuen KY. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan. Emerg Microbes Infect. 2020;9(1):221-236. [PMC free article: PMC7067204] [PubMed: 31987001]4.Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat Med. 2020 Apr;26(4):450-452. [PMC free article: PMC7095063] [PubMed: 32284615]5.Zhang T, Wu Q, Zhang Z. Probable Pangolin Origin of SARS-CoV-2 Associated with the COVID-19 Outbreak. Curr Biol. 2020 Apr 06;30(7):1346-1351.e2. [PMC free article: PMC7156161] [PubMed: 32197085]6.Oreshkova N, Molenaar RJ, Vreman S, Harders F, Oude Munnink BB, Hakze-van der Honing RW, Gerhards N, Tolsma P, Bouwstra R, Sikkema RS, Tacken MG, de Rooij MM, Weesendorp E, Engelsma MY, Bruschke CJ, Smit LA, Koopmans M, van der Poel WH, Stegeman A. SARS-CoV-2 infection in farmed minks, the Netherlands, April and May 2020. Euro Surveill. 2020 Jun;25(23) [PMC free article: PMC7403642] [PubMed: 32553059]7.Korber B, Fischer WM, Gnanakaran S, Yoon H, Theiler J, Abfalterer W, Hengartner N, Giorgi EE, Bhattacharya T, Foley B, Hastie KM, Parker MD, Partridge DG, Evans CM, Freeman TM, de Silva TI, Sheffield COVID-19 Genomics Group. McDanal C, Perez LG, Tang H, Moon-Walker A, Whelan SP, LaBranche CC, Saphire EO, Montefiori DC. Tracking Changes in SARS-CoV-2 Spike: Evidence that D614G Increases Infectivity of the COVID-19 Virus. Cell. 2020 Aug 20;182(4):812-827.e19. [PMC free article: PMC7332439] [PubMed: 32697968]8.Galloway SE, Paul P, MacCannell DR, Johansson MA, Brooks JT, MacNeil A, Slayton RB, Tong S, Silk BJ, Armstrong GL, Biggerstaff M, Dugan VG. Emergence of SARS-CoV-2 B.1.1.7 Lineage – United States, December 29, 2020-January 12, 2021. MMWR Morb Mortal Wkly Rep. 2021 Jan 22;70(3):95-99. [PMC free article: PMC7821772] [PubMed: 33476315]9.Volz E, Mishra S, Chand M, Barrett JC, Johnson R, Geidelberg L, Hinsley WR, Laydon DJ, Dabrera G, O\’Toole Á, Amato R, Ragonnet-Cronin M, Harrison I, Jackson B, Ariani CV, Boyd O, Loman NJ, McCrone JT, Gonçalves S, Jorgensen D, Myers R, Hill V, Jackson DK, Gaythorpe K, Groves N, Sillitoe J, Kwiatkowski DP, COVID-19 Genomics UK (COG-UK) consortium. Flaxman S, Ratmann O, Bhatt S, Hopkins S, Gandy A, Rambaut A, Ferguson NM. Assessing transmissibility of SARS-CoV-2 lineage B.1.1.7 in England. Nature. 2021 May;593(7858):266-269. [PubMed: 33767447]10.Wu K, Werner AP, Moliva JI, Koch M, Choi A, Stewart-Jones GBE, Bennett H, Boyoglu-Barnum S, Shi W, Graham BS, Carfi A, Corbett KS, Seder RA, Edwards DK. mRNA-1273 vaccine induces neutralizing antibodies against spike mutants from global SARS-CoV-2 variants. bioRxiv. 2021 Jan 25; [PubMed: 33501442]11.Davies NG, Abbott S, Barnard RC, Jarvis CI, Kucharski AJ, Munday JD, Pearson CAB, Russell TW, Tully DC, Washburne AD, Wenseleers T, Gimma A, Waites W, Wong KLM, van Zandvoort K, Silverman JD, CMMID COVID-19 Working Group. COVID-19 Genomics UK (COG-UK) Consortium. Diaz-Ordaz K, Keogh R, Eggo RM, Funk S, Jit M, Atkins KE, Edmunds WJ. Estimated transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science. 2021 Apr 09;372(6538) [PMC free article: PMC8128288] [PubMed: 33658326]12.Walensky RP, Walke HT, Fauci AS. SARS-CoV-2 Variants of Concern in the United States-Challenges and Opportunities. JAMA. 2021 Mar 16;325(11):1037-1038. [PubMed: 33595644]13.Davies NG, Barnard RC, Jarvis CI, Russell TW, Semple MG, Jit M, Edmunds WJ., Centre for Mathematical Modelling of Infectious Diseases COVID-19 Working Group. ISARIC4C investigators. Association of tiered restrictions and a second lockdown with COVID-19 deaths and hospital admissions in England: a modelling study. Lancet Infect Dis. 2021 Apr;21(4):482-492. [PMC free article: PMC7758181] [PubMed: 33357518]14.Challen R, Brooks-Pollock E, Read JM, Dyson L, Tsaneva-Atanasova K, Danon L. Risk of mortality in patients infected with SARS-CoV-2 variant of concern 202012/1: matched cohort study. BMJ. 2021 Mar 09;372:n579. [PMC free article: PMC7941603] [PubMed: 33687922]15.Davies NG, Jarvis CI, CMMID COVID-19 Working Group. Edmunds WJ, Jewell NP, Diaz-Ordaz K, Keogh RH. Increased mortality in community-tested cases of SARS-CoV-2 lineage B.1.1.7. Nature. 2021 May;593(7858):270-274. [PubMed: 33723411]16.Grint DJ, Wing K, Williamson E, McDonald HI, Bhaskaran K, Evans D, Evans SJ, Walker AJ, Hickman G, Nightingale E, Schultze A, Rentsch CT, Bates C, Cockburn J, Curtis HJ, Morton CE, Bacon S, Davy S, Wong AY, Mehrkar A, Tomlinson L, Douglas IJ, Mathur R, Blomquist P, MacKenna B, Ingelsby P, Croker R, Parry J, Hester F, Harper S, DeVito NJ, Hulme W, Tazare J, Goldacre B, Smeeth L, Eggo RM. Case fatality risk of the SARS-CoV-2 variant of concern B.1.1.7 in England, 16 November to 5 February. Euro Surveill. 2021 Mar;26(11) [PMC free article: PMC7976383] [PubMed: 33739254]17.Tegally H, Wilkinson E, Giovanetti M, Iranzadeh A, Fonseca V, Giandhari J, Doolabh D, Pillay S, San EJ, Msomi N, Mlisana K, von Gottberg A, Walaza S, Allam M, Ismail A, Mohale T, Glass AJ, Engelbrecht S, Van Zyl G, Preiser W, Petruccione F, Sigal A, Hardie D, Marais G, Hsiao NY, Korsman S, Davies MA, Tyers L, Mudau I, York D, Maslo C, Goedhals D, Abrahams S, Laguda-Akingba O, Alisoltani-Dehkordi A, Godzik A, Wibmer CK, Sewell BT, Lourenço J, Alcantara LCJ, Kosakovsky Pond SL, Weaver S, Martin D, Lessells RJ, Bhiman JN, Williamson C, de Oliveira T. Detection of a SARS-CoV-2 variant of concern in South Africa. Nature. 2021 Apr;592(7854):438-443. [PubMed: 33690265]18.Wibmer CK, Ayres F, Hermanus T, Madzivhandila M, Kgagudi P, Oosthuysen B, Lambson BE, de Oliveira T, Vermeulen M, van der Berg K, Rossouw T, Boswell M, Ueckermann V, Meiring S, von Gottberg A, Cohen C, Morris L, Bhiman JN, Moore PL. SARS-CoV-2 501Y.V2 escapes neutralization by South African COVID-19 donor plasma. bioRxiv. 2021 Jan 19; [PubMed: 33501446]19.Mwenda M, Saasa N, Sinyange N, Busby G, Chipimo PJ, Hendry J, Kapona O, Yingst S, Hines JZ, Minchella P, Simulundu E, Changula K, Nalubamba KS, Sawa H, Kajihara M, Yamagishi J, Kapin\’a M, Kapata N, Fwoloshi S, Zulu P, Mulenga LB, Agolory S, Mukonka V, Bridges DJ. Detection of B.1.351 SARS-CoV-2 Variant Strain – Zambia, December 2020. MMWR Morb Mortal Wkly Rep. 2021 Feb 26;70(8):280-282. [PubMed: 33630820]20.Wang P, Casner RG, Nair MS, Wang M, Yu J, Cerutti G, Liu L, Kwong PD, Huang Y, Shapiro L, Ho DD. Increased Resistance of SARS-CoV-2 Variant P.1 to Antibody Neutralization. bioRxiv. 2021 Mar 02; [PubMed: 33688656]21.Faria NR, Mellan TA, Whittaker C, Claro IM, Candido DDS, Mishra S, Crispim MAE, Sales FC, Hawryluk I, McCrone JT, Hulswit RJG, Franco LAM, Ramundo MS, de Jesus JG, Andrade PS, Coletti TM, Ferreira GM, Silva CAM, Manuli ER, Pereira RHM, Peixoto PS, Kraemer MU, Gaburo N, Camilo CDC, Hoeltgebaum H, Souza WM, Rocha EC, de Souza LM, de Pinho MC, Araujo LJT, Malta FSV, de Lima AB, Silva JDP, Zauli DAG, de S Ferreira AC, Schnekenberg RP, Laydon DJ, Walker PGT, Schlüter HM, Dos Santos ALP, Vidal MS, Del Caro VS, Filho RMF, Dos Santos HM, Aguiar RS, Modena JLP, Nelson B, Hay JA, Monod M, Miscouridou X, Coupland H, Sonabend R, Vollmer M, Gandy A, Suchard MA, Bowden TA, Pond SLK, Wu CH, Ratmann O, Ferguson NM, Dye C, Loman NJ, Lemey P, Rambaut A, Fraiji NA, Carvalho MDPSS, Pybus OG, Flaxman S, Bhatt S, Sabino EC. Genomics and epidemiology of a novel SARS-CoV-2 lineage in Manaus, Brazil. medRxiv. 2021 Mar 03; [PMC free article: PMC8139423] [PubMed: 33688664]22.Zhang W, Davis BD, Chen SS, Sincuir Martinez JM, Plummer JT, Vail E. Emergence of a Novel SARS-CoV-2 Variant in Southern California. JAMA. 2021 Apr 06;325(13):1324-1326. [PMC free article: PMC7879386] [PubMed: 33571356]23.van Doremalen N, Bushmaker T, Morris DH, Holbrook MG, Gamble A, Williamson BN, Tamin A, Harcourt JL, Thornburg NJ, Gerber SI, Lloyd-Smith JO, de Wit E, Munster VJ. Aerosol and Surface Stability of SARS-CoV-2 as Compared with SARS-CoV-1. N Engl J Med. 2020 Apr 16;382(16):1564-1567. [PMC free article: PMC7121658] [PubMed: 32182409]24.Riddell S, Goldie S, Hill A, Eagles D, Drew TW. The effect of temperature on persistence of SARS-CoV-2 on common surfaces. Virol J. 2020 Oct 07;17(1):145. [PMC free article: PMC7538848] [PubMed: 33028356]25.Guo ZD, Wang ZY, Zhang SF, Li X, Li L, Li C, Cui Y, Fu RB, Dong YZ, Chi XY, Zhang MY, Liu K, Cao C, Liu B, Zhang K, Gao YW, Lu B, Chen W. Aerosol and Surface Distribution of Severe Acute Respiratory Syndrome Coronavirus 2 in Hospital Wards, Wuhan, China, 2020. Emerg Infect Dis. 2020 Jul;26(7):1583-1591. [PMC free article: PMC7323510] [PubMed: 32275497]26.Yeo C, Kaushal S, Yeo D. Enteric involvement of coronaviruses: is faecal-oral transmission of SARS-CoV-2 possible? Lancet Gastroenterol Hepatol. 2020 Apr;5(4):335-337. [PMC free article: PMC7130008] [PubMed: 32087098]27.Kotlyar AM, Grechukhina O, Chen A, Popkhadze S, Grimshaw A, Tal O, Taylor HS, Tal R. Vertical transmission of coronavirus disease 2019: a systematic review and meta-analysis. Am J Obstet Gynecol. 2021 Jan;224(1):35-53.e3. [PMC free article: PMC7392880] [PubMed: 32739398]28.Ahmad FB, Cisewski JA, Miniño A, Anderson RN. Provisional Mortality Data – United States, 2020. MMWR Morb Mortal Wkly Rep. 2021 Apr 09;70(14):519-522. [PMC free article: PMC8030985] [PubMed: 33830988]29.Stokes EK, Zambrano LD, Anderson KN, Marder EP, Raz KM, El Burai Felix S, Tie Y, Fullerton KE. Coronavirus Disease 2019 Case Surveillance – United States, January 22-May 30, 2020. MMWR Morb Mortal Wkly Rep. 2020 Jun 19;69(24):759-765. [PMC free article: PMC7302472] [PubMed: 32555134]30.Gebhard C, Regitz-Zagrosek V, Neuhauser HK, Morgan R, Klein SL. Impact of sex and gender on COVID-19 outcomes in Europe. Biol Sex Differ. 2020 May 25;11(1):29. [PMC free article: PMC7247289] [PubMed: 32450906]31.Jin JM, Bai P, He W, Wu F, Liu XF, Han DM, Liu S, Yang JK. Gender Differences in Patients With COVID-19: Focus on Severity and Mortality. Front Public Health. 2020;8:152. [PMC free article: PMC7201103] [PubMed: 32411652]32.Finelli L, Gupta V, Petigara T, Yu K, Bauer KA, Puzniak LA. Mortality Among US Patients Hospitalized With SARS-CoV-2 Infection in 2020. JAMA Netw Open. 2021 Apr 01;4(4):e216556. [PMC free article: PMC8033442] [PubMed: 33830226]33.Romano SD, Blackstock AJ, Taylor EV, El Burai Felix S, Adjei S, Singleton CM, Fuld J, Bruce BB, Boehmer TK. Trends in Racial and Ethnic Disparities in COVID-19 Hospitalizations, by Region – United States, March-December 2020. MMWR Morb Mortal Wkly Rep. 2021 Apr 16;70(15):560-565. [PubMed: 33857068]34.Sze S, Pan D, Nevill CR, Gray LJ, Martin CA, Nazareth J, Minhas JS, Divall P, Khunti K, Abrams KR, Nellums LB, Pareek M. Ethnicity and clinical outcomes in COVID-19: A systematic review and meta-analysis. EClinicalMedicine. 2020 Dec;29:100630. [PMC free article: PMC7658622] [PubMed: 33200120]35.Heslin KC, Hall JE. Sexual Orientation Disparities in Risk Factors for Adverse COVID-19-Related Outcomes, by Race/Ethnicity – Behavioral Risk Factor Surveillance System, United States, 2017-2019. MMWR Morb Mortal Wkly Rep. 2021 Feb 05;70(5):149-154. [PMC free article: PMC7861482] [PubMed: 33539330]36.Song W, Gui M, Wang X, Xiang Y. Cryo-EM structure of the SARS coronavirus spike glycoprotein in complex with its host cell receptor ACE2. PLoS Pathog. 2018 Aug;14(8):e1007236. [PMC free article: PMC6107290] [PubMed: 30102747]37.Jiang S, Hillyer C, Du L. Neutralizing Antibodies against SARS-CoV-2 and Other Human Coronaviruses: (Trends in Immunology 41, 355-359; 2020). Trends Immunol. 2020 Jun;41(6):545. [PMC free article: PMC7271084] [PubMed: 32362491]38.Du L, He Y, Zhou Y, Liu S, Zheng BJ, Jiang S. The spike protein of SARS-CoV–a target for vaccine and therapeutic development. Nat Rev Microbiol. 2009 Mar;7(3):226-36. [PMC free article: PMC2750777] [PubMed: 19198616]39.Jiang S, Hillyer C, Du L. Neutralizing Antibodies against SARS-CoV-2 and Other Human Coronaviruses. Trends Immunol. 2020 May;41(5):355-359. [PMC free article: PMC7129017] [PubMed: 32249063]40.de Abajo FJ, Rodríguez-Martín S, Lerma V, Mejía-Abril G, Aguilar M, García-Luque A, Laredo L, Laosa O, Centeno-Soto GA, Ángeles Gálvez M, Puerro M, González-Rojano E, Pedraza L, de Pablo I, Abad-Santos F, Rodríguez-Mañas L, Gil M, Tobías A, Rodríguez-Miguel A, Rodríguez-Puyol D., MED-ACE2-COVID19 study group. Use of renin-angiotensin-aldosterone system inhibitors and risk of COVID-19 requiring admission to hospital: a case-population study. Lancet. 2020 May 30;395(10238):1705-1714. [PMC free article: PMC7255214] [PubMed: 32416785]41.Xu H, Zhong L, Deng J, Peng J, Dan H, Zeng X, Li T, Chen Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int J Oral Sci. 2020 Feb 24;12(1):8. [PMC free article: PMC7039956] [PubMed: 32094336]42.Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C, Pöhlmann S. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020 Apr 16;181(2):271-280.e8. [PMC free article: PMC7102627] [PubMed: 32142651]43.Wang J, Jiang M, Chen X, Montaner LJ. Cytokine storm and leukocyte changes in mild versus severe SARS-CoV-2 infection: Review of 3939 COVID-19 patients in China and emerging pathogenesis and therapy concepts. J Leukoc Biol. 2020 Jul;108(1):17-41. [PMC free article: PMC7323250] [PubMed: 32534467]44.Azkur AK, Akdis M, Azkur D, Sokolowska M, van de Veen W, Brüggen MC, O\’Mahony L, Gao Y, Nadeau K, Akdis CA. Immune response to SARS-CoV-2 and mechanisms of immunopathological changes in COVID-19. Allergy. 2020 Jul;75(7):1564-1581. [PMC free article: PMC7272948] [PubMed: 32396996]45.Teuwen LA, Geldhof V, Pasut A, Carmeliet P. COVID-19: the vasculature unleashed. Nat Rev Immunol. 2020 Jul;20(7):389-391. [PMC free article: PMC7240244] [PubMed: 32439870]46.Ackermann M, Verleden SE, Kuehnel M, Haverich A, Welte T, Laenger F, Vanstapel A, Werlein C, Stark H, Tzankov A, Li WW, Li VW, Mentzer SJ, Jonigk D. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N Engl J Med. 2020 Jul 09;383(2):120-128. [PMC free article: PMC7412750] [PubMed: 32437596]47.van de Veerdonk FL, Netea MG, van Deuren M, van der Meer JW, de Mast Q, Brüggemann RJ, van der Hoeven H. Kallikrein-kinin blockade in patients with COVID-19 to prevent acute respiratory distress syndrome. Elife. 2020 Apr 27;9 [PMC free article: PMC7213974] [PubMed: 32338605]48.Conti P, Ronconi G, Caraffa A, Gallenga CE, Ross R, Frydas I, Kritas SK. Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): anti-inflammatory strategies. 2020 March-April,J Biol Regul Homeost Agents. 34(2):327-331. [PubMed: 32171193]49.Coopersmith CM, Antonelli M, Bauer SR, Deutschman CS, Evans LE, Ferrer R, Hellman J, Jog S, Kesecioglu J, Kissoon N, Martin-Loeches I, Nunnally ME, Prescott HC, Rhodes A, Talmor D, Tissieres P, De Backer D. The Surviving Sepsis Campaign: Research Priorities for Coronavirus Disease 2019 in Critical Illness. Crit Care Med. 2021 Apr 01;49(4):598-622. [PubMed: 33591008]50.Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, Xu J, Gu X, Cheng Z, Yu T, Xia J, Wei Y, Wu W, Xie X, Yin W, Li H, Liu M, Xiao Y, Gao H, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020 Feb 15;395(10223):497-506. [PMC free article: PMC7159299] [PubMed: 31986264]51.Liu H, Wei P, Zhang Q, Chen Z, Aviszus K, Downing W, Peterson S, Reynoso L, Downey GP, Frankel SK, Kappler J, Marrack P, Zhang G. 501Y.V2 and 501Y.V3 variants of SARS-CoV-2 lose binding to Bamlanivimab in vitro. bioRxiv. 2021 Feb 16; [PubMed: 33619479]52.Hua A, O\’Gallagher K, Sado D, Byrne J. Life-threatening cardiac tamponade complicating myo-pericarditis in COVID-19. Eur Heart J. 2020 Jun 07;41(22):2130. [PMC free article: PMC7184427] [PubMed: 32227076]53.Guo T, Fan Y, Chen M, Wu X, Zhang L, He T, Wang H, Wan J, Wang X, Lu Z. Cardiovascular Implications of Fatal Outcomes of Patients With Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020 Jul 01;5(7):811-818. [PMC free article: PMC7101506] [PubMed: 32219356]54.Libby P, Loscalzo J, Ridker PM, Farkouh ME, Hsue PY, Fuster V, Hasan AA, Amar S. Inflammation, Immunity, and Infection in Atherothrombosis: JACC Review Topic of the Week. J Am Coll Cardiol. 2018 Oct 23;72(17):2071-2081. [PMC free article: PMC6196735] [PubMed: 30336831]55.Tan L, Wang Q, Zhang D, Ding J, Huang Q, Tang YQ, Wang Q, Miao H. Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study. Signal Transduct Target Ther. 2020 Mar 27;5(1):33. [PMC free article: PMC7100419] [PubMed: 32296069]56.Abou-Ismail MY, Diamond A, Kapoor S, Arafah Y, Nayak L. The hypercoagulable state in COVID-19: Incidence, pathophysiology, and management. Thromb Res. 2020 Oct;194:101-115. [PMC free article: PMC7305763] [PubMed: 32788101]57.Amgalan A, Othman M. Exploring possible mechanisms for COVID-19 induced thrombocytopenia: Unanswered questions. J Thromb Haemost. 2020 Jun;18(6):1514-1516. [PMC free article: PMC7262247] [PubMed: 32278338]58.Ni W, Yang X, Yang D, Bao J, Li R, Xiao Y, Hou C, Wang H, Liu J, Yang D, Xu Y, Cao Z, Gao Z. Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19. Crit Care. 2020 Jul 13;24(1):422. [PMC free article: PMC7356137] [PubMed: 32660650]59.Zubair AS, McAlpine LS, Gardin T, Farhadian S, Kuruvilla DE, Spudich S. Neuropathogenesis and Neurologic Manifestations of the Coronaviruses in the Age of Coronavirus Disease 2019: A Review. JAMA Neurol. 2020 Aug 01;77(8):1018-1027. [PMC free article: PMC7484225] [PubMed: 32469387]60.Patel KP, Patel PA, Vunnam RR, Hewlett AT, Jain R, Jing R, Vunnam SR. Gastrointestinal, hepatobiliary, and pancreatic manifestations of COVID-19. J Clin Virol. 2020 Jul;128:104386. [PMC free article: PMC7189193] [PubMed: 32388469]61.Gabarre P, Dumas G, Dupont T, Darmon M, Azoulay E, Zafrani L. Acute kidney injury in critically ill patients with COVID-19. Intensive Care Med. 2020 Jul;46(7):1339-1348. [PMC free article: PMC7290076] [PubMed: 32533197]62.Chi X, Yan R, Zhang J, Zhang G, Zhang Y, Hao M, Zhang Z, Fan P, Dong Y, Yang Y, Chen Z, Guo Y, Zhang J, Li Y, Song X, Chen Y, Xia L, Fu L, Hou L, Xu J, Yu C, Li J, Zhou Q, Chen W. A neutralizing human antibody binds to the N-terminal domain of the Spike protein of SARS-CoV-2. Science. 2020 Aug 07;369(6504):650-655. [PMC free article: PMC7319273] [PubMed: 32571838]63.Liu H, Zhang Q, Wei P, Chen Z, Aviszus K, Yang J, Downing W, Peterson S, Jiang C, Liang B, Reynoso L, Downey GP, Frankel SK, Kappler J, Marrack P, Zhang G. The basis of a more contagious 501Y.V1 variant of SARS-COV-2. bioRxiv. 2021 Feb 02; [PMC free article: PMC8063779] [PubMed: 33564771]64.Borczuk AC, Salvatore SP, Seshan SV, Patel SS, Bussel JB, Mostyka M, Elsoukkary S, He B, Del Vecchio C, Fortarezza F, Pezzuto F, Navalesi P, Crisanti A, Fowkes ME, Bryce CH, Calabrese F, Beasley MB. COVID-19 pulmonary pathology: a multi-institutional autopsy cohort from Italy and New York City. Mod Pathol. 2020 Nov;33(11):2156-2168. [PMC free article: PMC7463226] [PubMed: 32879413]65.Solomon IH, Normandin E, Bhattacharyya S, Mukerji SS, Keller K, Ali AS, Adams G, Hornick JL, Padera RF, Sabeti P. Neuropathological Features of Covid-19. N Engl J Med. 2020 Sep 03;383(10):989-992. [PMC free article: PMC7304421] [PubMed: 32530583]66.Lindner D, Fitzek A, Bräuninger H, Aleshcheva G, Edler C, Meissner K, Scherschel K, Kirchhof P, Escher F, Schultheiss HP, Blankenberg S, Püschel K, Westermann D. Association of Cardiac Infection With SARS-CoV-2 in Confirmed COVID-19 Autopsy Cases. JAMA Cardiol. 2020 Nov 01;5(11):1281-1285. [PMC free article: PMC7385672] [PubMed: 32730555]67.Su H, Yang M, Wan C, Yi LX, Tang F, Zhu HY, Yi F, Yang HC, Fogo AB, Nie X, Zhang C. Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney Int. 2020 Jul;98(1):219-227. [PMC free article: PMC7194105] [PubMed: 32327202]68.Lin L, Jiang X, Zhang Z, Huang S, Zhang Z, Fang Z, Gu Z, Gao L, Shi H, Mai L, Liu Y, Lin X, Lai R, Yan Z, Li X, Shan H. Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut. 2020 Jun;69(6):997-1001. [PMC free article: PMC7316116] [PubMed: 32241899]69.Lax SF, Skok K, Zechner P, Kessler HH, Kaufmann N, Koelblinger C, Vander K, Bargfrieder U, Trauner M. Pulmonary Arterial Thrombosis in COVID-19 With Fatal Outcome : Results From a Prospective, Single-Center, Clinicopathologic Case Series. Ann Intern Med. 2020 Sep 01;173(5):350-361. [PMC free article: PMC7249507] [PubMed: 32422076]70.Lauer SA, Grantz KH, Bi Q, Jones FK, Zheng Q, Meredith HR, Azman AS, Reich NG, Lessler J. The Incubation Period of Coronavirus Disease 2019 (COVID-19) From Publicly Reported Confirmed Cases: Estimation and Application. Ann Intern Med. 2020 May 05;172(9):577-582. [PMC free article: PMC7081172] [PubMed: 32150748]71.Mizumoto K, Kagaya K, Zarebski A, Chowell G. Estimating the asymptomatic proportion of coronavirus disease 2019 (COVID-19) cases on board the Diamond Princess cruise ship, Yokohama, Japan, 2020. Euro Surveill. 2020 Mar;25(10) [PMC free article: PMC7078829] [PubMed: 32183930]72.Nishiura H, Kobayashi T, Miyama T, Suzuki A, Jung SM, Hayashi K, Kinoshita R, Yang Y, Yuan B, Akhmetzhanov AR, Linton NM. Estimation of the asymptomatic ratio of novel coronavirus infections (COVID-19). Int J Infect Dis. 2020 May;94:154-155. [PMC free article: PMC7270890] [PubMed: 32179137]73.Zhu J, Zhong Z, Ji P, Li H, Li B, Pang J, Zhang J, Zhao C. Clinicopathological characteristics of 8697 patients with COVID-19 in China: a meta-analysis. Fam Med Community Health. 2020 Apr;8(2) [PMC free article: PMC7229787] [PubMed: 32371463]74.Li J, Huang DQ, Zou B, Yang H, Hui WZ, Rui F, Yee NTS, Liu C, Nerurkar SN, Kai JCY, Teng MLP, Li X, Zeng H, Borghi JA, Henry L, Cheung R, Nguyen MH. Epidemiology of COVID-19: A systematic review and meta-analysis of clinical characteristics, risk factors, and outcomes. J Med Virol. 2021 Mar;93(3):1449-1458. [PMC free article: PMC7436673] [PubMed: 32790106]75.Yang AP, Liu JP, Tao WQ, Li HM. The diagnostic and predictive role of NLR, d-NLR and PLR in COVID-19 patients. Int Immunopharmacol. 2020 Jul;84:106504. [PMC free article: PMC7152924] [PubMed: 32304994]76.ARDS Definition Task Force. Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012 Jun 20;307(23):2526-33. [PubMed: 22797452]77.Ferrando C, Suarez-Sipmann F, Mellado-Artigas R, Hernández M, Gea A, Arruti E, Aldecoa C, Martínez-Pallí G, Martínez-González MA, Slutsky AS, Villar J., COVID-19 Spanish ICU Network. Clinical features, ventilatory management, and outcome of ARDS caused by COVID-19 are similar to other causes of ARDS. Intensive Care Med. 2020 Dec;46(12):2200-2211. [PMC free article: PMC7387884] [PubMed: 32728965]78.Martinez-Rojas MA, Vega-Vega O, Bobadilla NA. Is the kidney a target of SARS-CoV-2? Am J Physiol Renal Physiol. 2020 Jun 01;318(6):F1454-F1462. [PMC free article: PMC7303722] [PubMed: 32412303]79.Hirsch JS, Ng JH, Ross DW, Sharma P, Shah HH, Barnett RL, Hazzan AD, Fishbane S, Jhaveri KD., Northwell COVID-19 Research Consortium. Northwell Nephrology COVID-19 Research Consortium. Acute kidney injury in patients hospitalized with COVID-19. Kidney Int. 2020 Jul;98(1):209-218. [PMC free article: PMC7229463] [PubMed: 32416116]80.Gupta A, Madhavan MV, Sehgal K, Nair N, Mahajan S, Sehrawat TS, Bikdeli B, Ahluwalia N, Ausiello JC, Wan EY, Freedberg DE, Kirtane AJ, Parikh SA, Maurer MS, Nordvig AS, Accili D, Bathon JM, Mohan S, Bauer KA, Leon MB, Krumholz HM, Uriel N, Mehra MR, Elkind MSV, Stone GW, Schwartz A, Ho DD, Bilezikian JP, Landry DW. Extrapulmonary manifestations of COVID-19. Nat Med. 2020 Jul;26(7):1017-1032. [PubMed: 32651579]81.Hessami A, Shamshirian A, Heydari K, Pourali F, Alizadeh-Navaei R, Moosazadeh M, Abrotan S, Shojaie L, Sedighi S, Shamshirian D, Rezaei N. Cardiovascular diseases burden in COVID-19: Systematic review and meta-analysis. Am J Emerg Med. 2020 Oct 16; [PMC free article: PMC7561581] [PubMed: 33268238]82.Tariq R, Saha S, Furqan F, Hassett L, Pardi D, Khanna S. Prevalence and Mortality of COVID-19 Patients With Gastrointestinal Symptoms: A Systematic Review and Meta-analysis. Mayo Clin Proc. 2020 Aug;95(8):1632-1648. [PMC free article: PMC7284248] [PubMed: 32753138]83.Azouz E, Yang S, Monnier-Cholley L, Arrivé L. Systemic arterial thrombosis and acute mesenteric ischemia in a patient with COVID-19. Intensive Care Med. 2020 Jul;46(7):1464-1465. [PMC free article: PMC7232609] [PubMed: 32424482]84.Xu L, Liu J, Lu M, Yang D, Zheng X. Liver injury during highly pathogenic human coronavirus infections. Liver Int. 2020 May;40(5):998-1004. [PMC free article: PMC7228361] [PubMed: 32170806]85.Toscano G, Palmerini F, Ravaglia S, Ruiz L, Invernizzi P, Cuzzoni MG, Franciotta D, Baldanti F, Daturi R, Postorino P, Cavallini A, Micieli G. Guillain-Barré Syndrome Associated with SARS-CoV-2. N Engl J Med. 2020 Jun 25;382(26):2574-2576. [PMC free article: PMC7182017] [PubMed: 32302082]86.Daneshgaran G, Dubin DP, Gould DJ. Cutaneous Manifestations of COVID-19: An Evidence-Based Review. Am J Clin Dermatol. 2020 Oct;21(5):627-639. [PMC free article: PMC7456663] [PubMed: 32865778]87.Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Pathophysiology, Transmission, Diagnosis, and Treatment of Coronavirus Disease 2019 (COVID-19): A Review. JAMA. 2020 Aug 25;324(8):782-793. [PubMed: 32648899]88.Gandhi RT, Lynch JB, Del Rio C. Mild or Moderate Covid-19. N Engl J Med. 2020 Oct 29;383(18):1757-1766. [PubMed: 32329974]89.Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, Shi Z, Hu Z, Zhong W, Xiao G. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 2020 Mar;30(3):269-271. [PMC free article: PMC7054408] [PubMed: 32020029]90.Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, Hohmann E, Chu HY, Luetkemeyer A, Kline S, Lopez de Castilla D, Finberg RW, Dierberg K, Tapson V, Hsieh L, Patterson TF, Paredes R, Sweeney DA, Short WR, Touloumi G, Lye DC, Ohmagari N, Oh MD, Ruiz-Palacios GM, Benfield T, Fätkenheuer G, Kortepeter MG, Atmar RL, Creech CB, Lundgren J, Babiker AG, Pett S, Neaton JD, Burgess TH, Bonnett T, Green M, Makowski M, Osinusi A, Nayak S, Lane HC., ACTT-1 Study Group Members. Remdesivir for the Treatment of Covid-19 – Final Report. N Engl J Med. 2020 Nov 05;383(19):1813-1826. [PMC free article: PMC7262788] [PubMed: 32445440]91.Goldman JD, Lye DCB, Hui DS, Marks KM, Bruno R, Montejano R, Spinner CD, Galli M, Ahn MY, Nahass RG, Chen YS, SenGupta D, Hyland RH, Osinusi AO, Cao H, Blair C, Wei X, Gaggar A, Brainard DM, Towner WJ, Muñoz J, Mullane KM, Marty FM, Tashima KT, Diaz G, Subramanian A., GS-US-540-5773 Investigators. Remdesivir for 5 or 10 Days in Patients with Severe Covid-19. N Engl J Med. 2020 Nov 05;383(19):1827-1837. [PMC free article: PMC7377062] [PubMed: 32459919]92.Spinner CD, Gottlieb RL, Criner GJ, Arribas López JR, Cattelan AM, Soriano Viladomiu A, Ogbuagu O, Malhotra P, Mullane KM, Castagna A, Chai LYA, Roestenberg M, Tsang OTY, Bernasconi E, Le Turnier P, Chang SC, SenGupta D, Hyland RH, Osinusi AO, Cao H, Blair C, Wang H, Gaggar A, Brainard DM, McPhail MJ, Bhagani S, Ahn MY, Sanyal AJ, Huhn G, Marty FM., GS-US-540-5774 Investigators. Effect of Remdesivir vs Standard Care on Clinical Status at 11 Days in Patients With Moderate COVID-19: A Randomized Clinical Trial. JAMA. 2020 Sep 15;324(11):1048-1057. [PMC free article: PMC7442954] [PubMed: 32821939]93.Zhang R, Mylonakis E. In inpatients with COVID-19, none of remdesivir, hydroxychloroquine, lopinavir, or interferon β-1a differed from standard care for in-hospital mortality. Ann Intern Med. 2021 Feb;174(2):JC17. [PubMed: 33524282]94.RECOVERY Collaborative Group. Horby P, Mafham M, Linsell L, Bell JL, Staplin N, Emberson JR, Wiselka M, Ustianowski A, Elmahi E, Prudon B, Whitehouse T, Felton T, Williams J, Faccenda J, Underwood J, Baillie JK, Chappell LC, Faust SN, Jaki T, Jeffery K, Lim WS, Montgomery A, Rowan K, Tarning J, Watson JA, White NJ, Juszczak E, Haynes R, Landray MJ. Effect of Hydroxychloroquine in Hospitalized Patients with Covid-19. N Engl J Med. 2020 Nov 19;383(21):2030-2040. [PMC free article: PMC7556338] [PubMed: 33031652]95.Mitjà O, Corbacho-Monné M, Ubals M, Alemany A, Suñer C, Tebé C, Tobias A, Peñafiel J, Ballana E, Pérez CA, Admella P, Riera-Martí N, Laporte P, Mitjà J, Clua M, Bertran L, Sarquella M, Gavilán S, Ara J, Argimon JM, Cuatrecasas G, Cañadas P, Elizalde-Torrent A, Fabregat R, Farré M, Forcada A, Flores-Mateo G, López C, Muntada E, Nadal N, Narejos S, Nieto A, Prat N, Puig J, Quiñones C, Ramírez-Viaplana F, Reyes-Urueña J, Riveira-Muñoz E, Ruiz L, Sanz S, Sentís A, Sierra A, Velasco C, Vivanco-Hidalgo RM, Zamora J, Casabona J, Vall-Mayans M, González-Beiras C, Clotet B., BCN-PEP-CoV2 Research Group. A Cluster-Randomized Trial of Hydroxychloroquine for Prevention of Covid-19. N Engl J Med. 2021 Feb 04;384(5):417-427. [PMC free article: PMC7722693] [PubMed: 33289973]96.Boulware DR, Pullen MF, Bangdiwala AS, Pastick KA, Lofgren SM, Okafor EC, Skipper CP, Nascene AA, Nicol MR, Abassi M, Engen NW, Cheng MP, LaBar D, Lother SA, MacKenzie LJ, Drobot G, Marten N, Zarychanski R, Kelly LE, Schwartz IS, McDonald EG, Rajasingham R, Lee TC, Hullsiek KH. A Randomized Trial of Hydroxychloroquine as Postexposure Prophylaxis for Covid-19. N Engl J Med. 2020 Aug 06;383(6):517-525. [PMC free article: PMC7289276] [PubMed: 32492293]97.Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G, Ruan L, Song B, Cai Y, Wei M, Li X, Xia J, Chen N, Xiang J, Yu T, Bai T, Xie X, Zhang L, Li C, Yuan Y, Chen H, Li H, Huang H, Tu S, Gong F, Liu Y, Wei Y, Dong C, Zhou F, Gu X, Xu J, Liu Z, Zhang Y, Li H, Shang L, Wang K, Li K, Zhou X, Dong X, Qu Z, Lu S, Hu X, Ruan S, Luo S, Wu J, Peng L, Cheng F, Pan L, Zou J, Jia C, Wang J, Liu X, Wang S, Wu X, Ge Q, He J, Zhan H, Qiu F, Guo L, Huang C, Jaki T, Hayden FG, Horby PW, Zhang D, Wang C. A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19. N Engl J Med. 2020 May 07;382(19):1787-1799. [PMC free article: PMC7121492] [PubMed: 32187464]98.Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Res. 2020 Jun;178:104787. [PMC free article: PMC7129059] [PubMed: 32251768]99.López-Medina E, López P, Hurtado IC, Dávalos DM, Ramirez O, Martínez E, Díazgranados JA, Oñate JM, Chavarriaga H, Herrera S, Parra B, Libreros G, Jaramillo R, Avendaño AC, Toro DF, Torres M, Lesmes MC, Rios CA, Caicedo I. Effect of Ivermectin on Time to Resolution of Symptoms Among Adults With Mild COVID-19: A Randomized Clinical Trial. JAMA. 2021 Apr 13;325(14):1426-1435. [PMC free article: PMC7934083] [PubMed: 33662102]100.Joyner MJ, Senefeld JW, Klassen SA, Mills JR, Johnson PW, Theel ES, Wiggins CC, Bruno KA, Klompas AM, Lesser ER, Kunze KL, Sexton MA, Diaz Soto JC, Baker SE, Shepherd JRA, van Helmond N, van Buskirk CM, Winters JL, Stubbs JR, Rea RF, Hodge DO, Herasevich V, Whelan ER, Clayburn AJ, Larson KF, Ripoll JG, Andersen KJ, Buras MR, Vogt MNP, Dennis JJ, Regimbal RJ, Bauer PR, Blair JE, Paneth NS, Fairweather D, Wright RS, Carter RE, Casadevall A. Effect of Convalescent Plasma on Mortality among Hospitalized Patients with COVID-19: Initial Three-Month Experience. medRxiv. 2020 Aug 12; [PubMed: 32817978]101.Joyner MJ, Bruno KA, Klassen SA, Kunze KL, Johnson PW, Lesser ER, Wiggins CC, Senefeld JW, Klompas AM, Hodge DO, Shepherd JRA, Rea RF, Whelan ER, Clayburn AJ, Spiegel MR, Baker SE, Larson KF, Ripoll JG, Andersen KJ, Buras MR, Vogt MNP, Herasevich V, Dennis JJ, Regimbal RJ, Bauer PR, Blair JE, van Buskirk CM, Winters JL, Stubbs JR, van Helmond N, Butterfield BP, Sexton MA, Diaz Soto JC, Paneth NS, Verdun NC, Marks P, Casadevall A, Fairweather D, Carter RE, Wright RS. Safety Update: COVID-19 Convalescent Plasma in 20,000 Hospitalized Patients. Mayo Clin Proc. 2020 Sep;95(9):1888-1897. [PMC free article: PMC7368917] [PubMed: 32861333]102.Simonovich VA, Burgos Pratx LD, Scibona P, Beruto MV, Vallone MG, Vázquez C, Savoy N, Giunta DH, Pérez LG, Sánchez MDL, Gamarnik AV, Ojeda DS, Santoro DM, Camino PJ, Antelo S, Rainero K, Vidiella GP, Miyazaki EA, Cornistein W, Trabadelo OA, Ross FM, Spotti M, Funtowicz G, Scordo WE, Losso MH, Ferniot I, Pardo PE, Rodriguez E, Rucci P, Pasquali J, Fuentes NA, Esperatti M, Speroni GA, Nannini EC, Matteaccio A, Michelangelo HG, Follmann D, Lane HC, Belloso WH., PlasmAr Study Group. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N Engl J Med. 2021 Feb 18;384(7):619-629. [PMC free article: PMC7722692] [PubMed: 33232588]103.Li L, Zhang W, Hu Y, Tong X, Zheng S, Yang J, Kong Y, Ren L, Wei Q, Mei H, Hu C, Tao C, Yang R, Wang J, Yu Y, Guo Y, Wu X, Xu Z, Zeng L, Xiong N, Chen L, Wang J, Man N, Liu Y, Xu H, Deng E, Zhang X, Li C, Wang C, Su S, Zhang L, Wang J, Wu Y, Liu Z. Effect of Convalescent Plasma Therapy on Time to Clinical Improvement in Patients With Severe and Life-threatening COVID-19: A Randomized Clinical Trial. JAMA. 2020 Aug 04;324(5):460-470. [PMC free article: PMC7270883] [PubMed: 32492084]104.Convalescent plasma in the management of moderate covid-19 in adults in India: open label phase II multicentre randomised controlled trial (PLACID Trial). BMJ. 2020 Nov 03;371:m4232. [PMC free article: PMC7607867] [PubMed: 33144278]105.Wibmer CK, Ayres F, Hermanus T, Madzivhandila M, Kgagudi P, Oosthuysen B, Lambson BE, de Oliveira T, Vermeulen M, van der Berg K, Rossouw T, Boswell M, Ueckermann V, Meiring S, von Gottberg A, Cohen C, Morris L, Bhiman JN, Moore PL. SARS-CoV-2 501Y.V2 escapes neutralization by South African COVID-19 donor plasma. Nat Med. 2021 Apr;27(4):622-625. [PubMed: 33654292]106.Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, Wang M, Yu J, Zhang B, Kwong PD, Graham BS, Mascola JR, Chang JY, Yin MT, Sobieszczyk M, Kyratsous CA, Shapiro L, Sheng Z, Huang Y, Ho DD. Antibody resistance of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature. 2021 May;593(7857):130-135. [PubMed: 33684923]107.Baum A, Ajithdoss D, Copin R, Zhou A, Lanza K, Negron N, Ni M, Wei Y, Mohammadi K, Musser B, Atwal GS, Oyejide A, Goez-Gazi Y, Dutton J, Clemmons E, Staples HM, Bartley C, Klaffke B, Alfson K, Gazi M, Gonzalez O, Dick E, Carrion R, Pessaint L, Porto M, Cook A, Brown R, Ali V, Greenhouse J, Taylor T, Andersen H, Lewis MG, Stahl N, Murphy AJ, Yancopoulos GD, Kyratsous CA. REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters. Science. 2020 Nov 27;370(6520):1110-1115. [PMC free article: PMC7857396] [PubMed: 33037066]108.Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, Musser BJ, Soo Y, Rofail D, Im J, Perry C, Pan C, Hosain R, Mahmood A, Davis JD, Turner KC, Hooper AT, Hamilton JD, Baum A, Kyratsous CA, Kim Y, Cook A, Kampman W, Kohli A, Sachdeva Y, Graber X, Kowal B, DiCioccio T, Stahl N, Lipsich L, Braunstein N, Herman G, Yancopoulos GD., Trial Investigators. REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19. N Engl J Med. 2021 Jan 21;384(3):238-251. [PMC free article: PMC7781102] [PubMed: 33332778]109.Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B, Morris J, Huhn G, Cardona J, Mocherla B, Stosor V, Shawa I, Kumar P, Adams AC, Van Naarden J, Custer KL, Durante M, Oakley G, Schade AE, Holzer TR, Ebert PJ, Higgs RE, Kallewaard NL, Sabo J, Patel DR, Klekotka P, Shen L, Skovronsky DM. Effect of Bamlanivimab as Monotherapy or in Combination With Etesevimab on Viral Load in Patients With Mild to Moderate COVID-19: A Randomized Clinical Trial. JAMA. 2021 Feb 16;325(7):632-644. [PMC free article: PMC7821080] [PubMed: 33475701]110.Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, Wang M, Yu J, Zhang B, Kwong PD, Graham BS, Mascola JR, Chang JY, Yin MT, Sobieszczyk M, Kyratsous CA, Shapiro L, Sheng Z, Huang Y, Ho DD. Increased Resistance of SARS-CoV-2 Variants B.1.351 and B.1.1.7 to Antibody Neutralization. bioRxiv. 2021 Jan 26; [PubMed: 33532778]111.RECOVERY Collaborative Group. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, Staplin N, Brightling C, Ustianowski A, Elmahi E, Prudon B, Green C, Felton T, Chadwick D, Rege K, Fegan C, Chappell LC, Faust SN, Jaki T, Jeffery K, Montgomery A, Rowan K, Juszczak E, Baillie JK, Haynes R, Landray MJ. Dexamethasone in Hospitalized Patients with Covid-19. N Engl J Med. 2021 Feb 25;384(8):693-704. [PMC free article: PMC7383595] [PubMed: 32678530]112.Yuen CK, Lam JY, Wong WM, Mak LF, Wang X, Chu H, Cai JP, Jin DY, To KK, Chan JF, Yuen KY, Kok KH. SARS-CoV-2 nsp13, nsp14, nsp15 and orf6 function as potent interferon antagonists. Emerg Microbes Infect. 2020 Dec;9(1):1418-1428. [PMC free article: PMC7473193] [PubMed: 32529952]113.Ranieri VM, Pettilä V, Karvonen MK, Jalkanen J, Nightingale P, Brealey D, Mancebo J, Ferrer R, Mercat A, Patroniti N, Quintel M, Vincent JL, Okkonen M, Meziani F, Bellani G, MacCallum N, Creteur J, Kluge S, Artigas-Raventos A, Maksimow M, Piippo I, Elima K, Jalkanen S, Jalkanen M, Bellingan G., INTEREST Study Group. Effect of Intravenous Interferon β-1a on Death and Days Free From Mechanical Ventilation Among Patients With Moderate to Severe Acute Respiratory Distress Syndrome: A Randomized Clinical Trial. JAMA. 2020 Feb 25;323(8):725-733. [PubMed: 32065831]114.Monk PD, Marsden RJ, Tear VJ, Brookes J, Batten TN, Mankowski M, Gabbay FJ, Davies DE, Holgate ST, Ho LP, Clark T, Djukanovic R, Wilkinson TMA., Inhaled Interferon Beta COVID-19 Study Group. Safety and efficacy of inhaled nebulised interferon beta-1a (SNG001) for treatment of SARS-CoV-2 infection: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Respir Med. 2021 Feb;9(2):196-206. [PMC free article: PMC7836724] [PubMed: 33189161]115.Davoudi-Monfared E, Rahmani H, Khalili H, Hajiabdolbaghi M, Salehi M, Abbasian L, Kazemzadeh H, Yekaninejad MS. A Randomized Clinical Trial of the Efficacy and Safety of Interferon β-1a in Treatment of Severe COVID-19. Antimicrob Agents Chemother. 2020 Aug 20;64(9) [PMC free article: PMC7449227] [PubMed: 32661006]116.Huet T, Beaussier H, Voisin O, Jouveshomme S, Dauriat G, Lazareth I, Sacco E, Naccache JM, Bézie Y, Laplanche S, Le Berre A, Le Pavec J, Salmeron S, Emmerich J, Mourad JJ, Chatellier G, Hayem G. Anakinra for severe forms of COVID-19: a cohort study. Lancet Rheumatol. 2020 Jul;2(7):e393-e400. [PMC free article: PMC7259909] [PubMed: 32835245]117.Cellina M, Orsi M, Bombaci F, Sala M, Marino P, Oliva G. Favorable changes of CT findings in a patient with COVID-19 pneumonia after treatment with tocilizumab. Diagn Interv Imaging. 2020 May;101(5):323-324. [PMC free article: PMC7270926] [PubMed: 32278585]118.Michot JM, Albiges L, Chaput N, Saada V, Pommeret F, Griscelli F, Balleyguier C, Besse B, Marabelle A, Netzer F, Merad M, Robert C, Barlesi F, Gachot B, Stoclin A. Tocilizumab, an anti-IL-6 receptor antibody, to treat COVID-19-related respiratory failure: a case report. Ann Oncol. 2020 Jul;31(7):961-964. [PMC free article: PMC7136869] [PubMed: 32247642]119.Rosas IO, Bräu N, Waters M, Go RC, Hunter BD, Bhagani S, Skiest D, Aziz MS, Cooper N, Douglas IS, Savic S, Youngstein T, Del Sorbo L, Cubillo Gracian A, De La Zerda DJ, Ustianowski A, Bao M, Dimonaco S, Graham E, Matharu B, Spotswood H, Tsai L, Malhotra A. Tocilizumab in Hospitalized Patients with Severe Covid-19 Pneumonia. N Engl J Med. 2021 Apr 22;384(16):1503-1516. [PMC free article: PMC7953459] [PubMed: 33631066]120.Stone JH, Frigault MJ, Serling-Boyd NJ, Fernandes AD, Harvey L, Foulkes AS, Horick NK, Healy BC, Shah R, Bensaci AM, Woolley AE, Nikiforow S, Lin N, Sagar M, Schrager H, Huckins DS, Axelrod M, Pincus MD, Fleisher J, Sacks CA, Dougan M, North CM, Halvorsen YD, Thurber TK, Dagher Z, Scherer A, Wallwork RS, Kim AY, Schoenfeld S, Sen P, Neilan TG, Perugino CA, Unizony SH, Collier DS, Matza MA, Yinh JM, Bowman KA, Meyerowitz E, Zafar A, Drobni ZD, Bolster MB, Kohler M, D\’Silva KM, Dau J, Lockwood MM, Cubbison C, Weber BN, Mansour MK., BACC Bay Tocilizumab Trial Investigators. Efficacy of Tocilizumab in Patients Hospitalized with Covid-19. N Engl J Med. 2020 Dec 10;383(24):2333-2344. [PMC free article: PMC7646626] [PubMed: 33085857]121.REMAP-CAP Investigators. Gordon AC, Mouncey PR, Al-Beidh F, Rowan KM, Nichol AD, Arabi YM, Annane D, Beane A, van Bentum-Puijk W, Berry LR, Bhimani Z, Bonten MJM, Bradbury CA, Brunkhorst FM, Buzgau A, Cheng AC, Detry MA, Duffy EJ, Estcourt LJ, Fitzgerald M, Goossens H, Haniffa R, Higgins AM, Hills TE, Horvat CM, Lamontagne F, Lawler PR, Leavis HL, Linstrum KM, Litton E, Lorenzi E, Marshall JC, Mayr FB, McAuley DF, McGlothlin A, McGuinness SP, McVerry BJ, Montgomery SK, Morpeth SC, Murthy S, Orr K, Parke RL, Parker JC, Patanwala AE, Pettilä V, Rademaker E, Santos MS, Saunders CT, Seymour CW, Shankar-Hari M, Sligl WI, Turgeon AF, Turner AM, van de Veerdonk FL, Zarychanski R, Green C, Lewis RJ, Angus DC, McArthur CJ, Berry S, Webb SA, Derde LPG. Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19. N Engl J Med. 2021 Apr 22;384(16):1491-1502. [PMC free article: PMC7953461] [PubMed: 33631065]122.Lescure FX, Honda H, Fowler RA, Lazar JS, Shi G, Wung P, Patel N, Hagino O., Sarilumab COVID-19 Global Study Group. Sarilumab in patients admitted to hospital with severe or critical COVID-19: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2021 May;9(5):522-532. [PMC free article: PMC8078879] [PubMed: 33676590]123.Richardson P, Griffin I, Tucker C, Smith D, Oechsle O, Phelan A, Rawling M, Savory E, Stebbing J. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet. 2020 Feb 15;395(10223):e30-e31. [PMC free article: PMC7137985] [PubMed: 32032529]124.Stebbing J, Phelan A, Griffin I, Tucker C, Oechsle O, Smith D, Richardson P. COVID-19: combining antiviral and anti-inflammatory treatments. Lancet Infect Dis. 2020 Apr;20(4):400-402. [PMC free article: PMC7158903] [PubMed: 32113509]125.Kalil AC, Patterson TF, Mehta AK, Tomashek KM, Wolfe CR, Ghazaryan V, Marconi VC, Ruiz-Palacios GM, Hsieh L, Kline S, Tapson V, Iovine NM, Jain MK, Sweeney DA, El Sahly HM, Branche AR, Regalado Pineda J, Lye DC, Sandkovsky U, Luetkemeyer AF, Cohen SH, Finberg RW, Jackson PEH, Taiwo B, Paules CI, Arguinchona H, Erdmann N, Ahuja N, Frank M, Oh MD, Kim ES, Tan SY, Mularski RA, Nielsen H, Ponce PO, Taylor BS, Larson L, Rouphael NG, Saklawi Y, Cantos VD, Ko ER, Engemann JJ, Amin AN, Watanabe M, Billings J, Elie MC, Davey RT, Burgess TH, Ferreira J, Green M, Makowski M, Cardoso A, de Bono S, Bonnett T, Proschan M, Deye GA, Dempsey W, Nayak SU, Dodd LE, Beigel JH., ACTT-2 Study Group Members. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. N Engl J Med. 2021 Mar 04;384(9):795-807. [PMC free article: PMC7745180] [PubMed: 33306283]126.Cao Y, Wei J, Zou L, Jiang T, Wang G, Chen L, Huang L, Meng F, Huang L, Wang N, Zhou X, Luo H, Mao Z, Chen X, Xie J, Liu J, Cheng H, Zhao J, Huang G, Wang W, Zhou J. Ruxolitinib in treatment of severe coronavirus disease 2019 (COVID-19): A multicenter, single-blind, randomized controlled trial. J Allergy Clin Immunol. 2020 Jul;146(1):137-146.e3. [PMC free article: PMC7250105] [PubMed: 32470486]127.Roschewski M, Lionakis MS, Sharman JP, Roswarski J, Goy A, Monticelli MA, Roshon M, Wrzesinski SH, Desai JV, Zarakas MA, Collen J, Rose K, Hamdy A, Izumi R, Wright GW, Chung KK, Baselga J, Staudt LM, Wilson WH. Inhibition of Bruton tyrosine kinase in patients with severe COVID-19. Sci Immunol. 2020 Jun 05;5(48) [PMC free article: PMC7274761] [PubMed: 32503877]128.Ni YN, Luo J, Yu H, Liu D, Liang BM, Liang ZA. The effect of high-flow nasal cannula in reducing the mortality and the rate of endotracheal intubation when used before mechanical ventilation compared with conventional oxygen therapy and noninvasive positive pressure ventilation. A systematic review and meta-analysis. Am J Emerg Med. 2018 Feb;36(2):226-233. [PubMed: 28780231]129.Hui DS, Chow BK, Lo T, Tsang OTY, Ko FW, Ng SS, Gin T, Chan MTV. Exhaled air dispersion during high-flow nasal cannula therapy versus CPAP via different masks. Eur Respir J. 2019 Apr;53(4) [PubMed: 30705129]130.Berlin DA, Gulick RM, Martinez FJ. Severe Covid-19. N Engl J Med. 2020 Dec 17;383(25):2451-2460. [PubMed: 32412710]131.Cook TM, El-Boghdadly K, McGuire B, McNarry AF, Patel A, Higgs A. Consensus guidelines for managing the airway in patients with COVID-19: Guidelines from the Difficult Airway Society, the Association of Anaesthetists the Intensive Care Society, the Faculty of Intensive Care Medicine and the Royal College of Anaesthetists. Anaesthesia. 2020 Jun;75(6):785-799. [PMC free article: PMC7383579] [PubMed: 32221970]132.Alhazzani W, Møller MH, Arabi YM, Loeb M, Gong MN, Fan E, Oczkowski S, Levy MM, Derde L, Dzierba A, Du B, Aboodi M, Wunsch H, Cecconi M, Koh Y, Chertow DS, Maitland K, Alshamsi F, Belley-Cote E, Greco M, Laundy M, Morgan JS, Kesecioglu J, McGeer A, Mermel L, Mammen MJ, Alexander PE, Arrington A, Centofanti JE, Citerio G, Baw B, Memish ZA, Hammond N, Hayden FG, Evans L, Rhodes A. Surviving Sepsis Campaign: Guidelines on the Management of Critically Ill Adults with Coronavirus Disease 2019 (COVID-19). Crit Care Med. 2020 Jun;48(6):e440-e469. [PMC free article: PMC7176264] [PubMed: 32224769]133.Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, Perez JL, Pérez Marc G, Moreira ED, Zerbini C, Bailey R, Swanson KA, Roychoudhury S, Koury K, Li P, Kalina WV, Cooper D, Frenck RW, Hammitt LL, Türeci Ö, Nell H, Schaefer A, Ünal S, Tresnan DB, Mather S, Dormitzer PR, Şahin U, Jansen KU, Gruber WC., C4591001 Clinical Trial Group. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N Engl J Med. 2020 Dec 31;383(27):2603-2615. [PMC free article: PMC7745181] [PubMed: 33301246]134.Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, Diemert D, Spector SA, Rouphael N, Creech CB, McGettigan J, Khetan S, Segall N, Solis J, Brosz A, Fierro C, Schwartz H, Neuzil K, Corey L, Gilbert P, Janes H, Follmann D, Marovich M, Mascola J, Polakowski L, Ledgerwood J, Graham BS, Bennett H, Pajon R, Knightly C, Leav B, Deng W, Zhou H, Han S, Ivarsson M, Miller J, Zaks T., COVE Study Group. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N Engl J Med. 2021 Feb 04;384(5):403-416. [PMC free article: PMC7787219] [PubMed: 33378609]135.Voysey M, Clemens SAC, Madhi SA, Weckx LY, Folegatti PM, Aley PK, Angus B, Baillie VL, Barnabas SL, Bhorat QE, Bibi S, Briner C, Cicconi P, Collins AM, Colin-Jones R, Cutland CL, Darton TC, Dheda K, Duncan CJA, Emary KRW, Ewer KJ, Fairlie L, Faust SN, Feng S, Ferreira DM, Finn A, Goodman AL, Green CM, Green CA, Heath PT, Hill C, Hill H, Hirsch I, Hodgson SHC, Izu A, Jackson S, Jenkin D, Joe CCD, Kerridge S, Koen A, Kwatra G, Lazarus R, Lawrie AM, Lelliott A, Libri V, Lillie PJ, Mallory R, Mendes AVA, Milan EP, Minassian AM, McGregor A, Morrison H, Mujadidi YF, Nana A, O\’Reilly PJ, Padayachee SD, Pittella A, Plested E, Pollock KM, Ramasamy MN, Rhead S, Schwarzbold AV, Singh N, Smith A, Song R, Snape MD, Sprinz E, Sutherland RK, Tarrant R, Thomson EC, Török ME, Toshner M, Turner DPJ, Vekemans J, Villafana TL, Watson MEE, Williams CJ, Douglas AD, Hill AVS, Lambe T, Gilbert SC, Pollard AJ., Oxford COVID Vaccine Trial Group. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet. 2021 Jan 09;397(10269):99-111. [PMC free article: PMC7723445] [PubMed: 33306989]136.Liu Y, Liu J, Xia H, Zhang X, Fontes-Garfias CR, Swanson KA, Cai H, Sarkar R, Chen W, Cutler M, Cooper D, Weaver SC, Muik A, Sahin U, Jansen KU, Xie X, Dormitzer PR, Shi PY. Neutralizing Activity of BNT162b2-Elicited Serum. N Engl J Med. 2021 Apr 15;384(15):1466-1468. [PMC free article: PMC7944950] [PubMed: 33684280]137.Wu K, Werner AP, Koch M, Choi A, Narayanan E, Stewart-Jones GBE, Colpitts T, Bennett H, Boyoglu-Barnum S, Shi W, Moliva JI, Sullivan NJ, Graham BS, Carfi A, Corbett KS, Seder RA, Edwards DK. Serum Neutralizing Activity Elicited by mRNA-1273 Vaccine. N Engl J Med. 2021 Apr 15;384(15):1468-1470. [PMC free article: PMC8008744] [PubMed: 33730471]138.Abdool Karim SS, de Oliveira T. New SARS-CoV-2 Variants – Clinical, Public Health, and Vaccine Implications. N Engl J Med. 2021 May 13;384(19):1866-1868. [PMC free article: PMC8008749] [PubMed: 33761203]139.Emary KRW, Golubchik T, Aley PK, Ariani CV, Angus B, Bibi S, Blane B, Bonsall D, Cicconi P, Charlton S, Clutterbuck EA, Collins AM, Cox T, Darton TC, Dold C, Douglas AD, Duncan CJA, Ewer KJ, Flaxman AL, Faust SN, Ferreira DM, Feng S, Finn A, Folegatti PM, Fuskova M, Galiza E, Goodman AL, Green CM, Green CA, Greenland M, Hallis B, Heath PT, Hay J, Hill HC, Jenkin D, Kerridge S, Lazarus R, Libri V, Lillie PJ, Ludden C, Marchevsky NG, Minassian AM, McGregor AC, Mujadidi YF, Phillips DJ, Plested E, Pollock KM, Robinson H, Smith A, Song R, Snape MD, Sutherland RK, Thomson EC, Toshner M, Turner DPJ, Vekemans J, Villafana TL, Williams CJ, Hill AVS, Lambe T, Gilbert SC, Voysey M, Ramasamy MN, Pollard AJ., COVID-19 Genomics UK consortium. AMPHEUS Project. Oxford COVID-19 Vaccine Trial Group. Efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B.1.1.7): an exploratory analysis of a randomised controlled trial. Lancet. 2021 Apr 10;397(10282):1351-1362. [PMC free article: PMC8009612] [PubMed: 33798499]140.COVID-ICU Group on behalf of the REVA Network and the COVID-ICU Investigators. Clinical characteristics and day-90 outcomes of 4244 critically ill adults with COVID-19: a prospective cohort study. Intensive Care Med. 2021 Jan;47(1):60-73. [PMC free article: PMC7674575] [PubMed: 33211135]141.Kaafarani HMA, El Moheb M, Hwabejire JO, Naar L, Christensen MA, Breen K, Gaitanidis A, Alser O, Mashbari H, Bankhead-Kendall B, Mokhtari A, Maurer L, Kapoen C, Langeveld K, El Hechi MW, Lee J, Mendoza AE, Saillant NN, Parks J, Fawley J, King DR, Fagenholz PJ, Velmahos GC. Gastrointestinal Complications in Critically Ill Patients With COVID-19. Ann Surg. 2020 Aug;272(2):e61-e62. [PMC free article: PMC7268843] [PubMed: 32675498]142.Zhou X, Cheng Z, Luo L, Zhu Y, Lin W, Ming Z, Chen W, Hu Y. Incidence and impact of disseminated intravascular coagulation in COVID-19 a systematic review and meta-analysis. Thromb Res. 2021 May;201:23-29. [PMC free article: PMC7886625] [PubMed: 33631519]143.Huang C, Huang L, Wang Y, Li X, Ren L, Gu X, Kang L, Guo L, Liu M, Zhou X, Luo J, Huang Z, Tu S, Zhao Y, Chen L, Xu D, Li Y, Li C, Peng L, Li Y, Xie W, Cui D, Shang L, Fan G, Xu J, Wang G, Wang Y, Zhong J, Wang C, Wang J, Zhang D, Cao B. 6-month consequences of COVID-19 in patients discharged from hospital: a cohort study. Lancet. 2021 Jan 16;397(10270):220-232. [PMC free article: PMC7833295] [PubMed: 33428867]144.Taquet M, Geddes JR, Husain M, Luciano S, Harrison PJ. 6-month neurological and psychiatric outcomes in 236 379 survivors of COVID-19: a retrospective cohort study using electronic health records. Lancet Psychiatry. 2021 May;8(5):416-427. [PMC free article: PMC8023694] [PubMed: 33836148]
Source