Next Article in Journal
Regulatory miRNA–mRNA Networks in Parkinson’s Disease
Previous Article in Journal
Targeting HDACs in Pancreatic Neuroendocrine Tumor Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Towards Goals to Refine Prophylactic and Therapeutic Strategies Against COVID-19 Linked to Aging and Metabolic Syndrome

Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
*
Authors to whom correspondence should be addressed.
Submission received: 22 April 2021 / Revised: 28 May 2021 / Accepted: 3 June 2021 / Published: 6 June 2021

Abstract

:
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) gave rise to the coronavirus disease 2019 (COVID-19) pandemic. A strong correlation has been demonstrated between worse COVID-19 outcomes, aging, and metabolic syndrome (MetS), which is primarily derived from obesity-induced systemic chronic low-grade inflammation with numerous complications, including type 2 diabetes mellitus (T2DM). The majority of COVID-19 deaths occurs in people over the age of 65. Individuals with MetS are inclined to manifest adverse disease consequences and mortality from COVID-19. In this review, we examine the prevalence and molecular mechanisms underlying enhanced risk of COVID-19 in elderly people and individuals with MetS. Subsequently, we discuss current progresses in treating COVID-19, including the development of new COVID-19 vaccines and antivirals, towards goals to elaborate prophylactic and therapeutic treatment options in this vulnerable population.

1. Introduction

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) brought about the coronavirus disease 2019 (COVID-19) pandemic causing an unprecedented crisis worldwide. SARS-CoV-2 is conveyed primarily through respiratory droplets [1]; SARS-CoV-2 enters the host airway epithelial cells via binding to the receptor, angiotensin-converting enzyme 2 (ACE2) [2]. While SARS-CoV-2 leads to a mild to moderate symptoms in most people, it provokes severe complications, including pneumonia, acute respiratory distress syndrome (ARDS), acute kidney injury, and organ failure with subsequent mortality, in older individuals and people with underlying medical conditions of heart/lung disease or diabetes mellitus (DM).
Aging leads to a loss of physiological integrity, functional decline, and increased vulnerability to death [3]. The prevalent feature of aging and age-related diseases is chronic inflammation, known as “inflammaging”; it is the low-grade, systemic inflammation without overt infection, and is a key risk factor for morbidity and mortality in older individuals [4]. The major contributing factors of inflammaging are: (1) endogenous host-derived cell debris that accumulate with age [5]; (2) senescent cells and their senescence-associated secretory phenotype (SASP) [6]; (3) immunosenescence exacerbated by continuing infections of viruses, leading to mild hyperactivity of innate immunity [7,8]; (4) gut- and other microbiota-derived harmful products and metabolites [9]; and (5) increasing activation of coagulation system with aging [5]. Specifically, constant activation of intrinsic receptors by endogenous signals has been suggested to direct a chronic state of background inflammation [7,8]. Cellular senescence and the attainment of the SASP by fibroblasts, endothelial, and immune cells have also been suggested to bestow inflammaging; cell senescence triggers the deposition of dysfunctional, terminally differentiated B, T, and natural killer (NK) cells [6]. Notably, inflammaging mechanistically connects aging/age-associated diseases with metabolic syndrome (MetS) [10].
MetS is a constellation of conditions, such as abdominal obesity, high blood pressure, insulin resistance (IR)/glucose intolerance, atherogenic dyslipidemia, pro-thrombotic state, and pro-inflammatory state; the likelihood of heart disease, stroke, and type 2 diabetes mellitus (T2DM) is increased by MetS [11]. Clinically, a diagnosis of MetS is determined by the presence of 3 or more of the following factors: increased waist circumference (>102 cm (>40 in) in men; >88 cm (>35 in) in women), hypertriglyceridemia (≥150 mg/dL), increased blood pressure (systolic ≥ 130 and/or diastolic ≥ 85 mmHg), decreased high-density lipoprotein (HDL) cholesterol (<40 mg/dL in males; <50 mg/dL in females), and dysglycemia (≥100 mg/dL) (Adult Treatment Panel III report (ATP III), National Cholesterol Education Program) [11].
The obesity epidemic has been crucially noticed as over 1.9 billion people being overweight with the worldwide prevalence of obesity nearly tripled since 1975 [12]. Obesity increases the risk of premature death by 1.45- to 2.76-fold and shortens lifespan by up to 20 years [13,14]. Obesity-induced systemic chronic low-grade inflammation is an essential mechanistic cause of MetS [12]. It is distinct from other inflammatory prototypes as it involves in continuous activation of the innate immune system, leading to tissue remodeling and systemic metabolic deterioration over time; it also sets off maladaptive reactions, resulting in substantial tissue damage. Multiple organs, including brain, heart, insulin-sensitive tissues (adipose, liver, and skeletal muscle), and pancreas are sites of inflammation, disrupting glucose and energy metabolism [12]. Accordingly, obesity and aging share a similar spectrum of phenotypes and mechanisms, such as redox imbalance, mitochondrial dysfunction, accumulation of macromolecules, weakened immunity, and systemic inflammation, and are sources of age-related diseases [15]. Unsurprisingly, conditions and comorbidities of aging and age-related diseases mirror those of obesity [15].
The essential influence of aging and MetS on viral infection and host susceptibility has been an important research focus [16,17,18,19]. Viruses can reprogram the host cell metabolism to allow their replication and progeny release while averting host immune responses to evade pathogen recognition. Because aging and metabolic disorders debilitate immune system, viral infection further impairs immune responses and enhances severity of metabolic disease [16,17,18,19]. Consequently, elderly people and patients with MetS often experience worse progression of viral diseases, such as unparalleled severe outcomes of COVID-19 (over 3 million death worldwide).
In this review, we examine the prevalence and molecular mechanisms underlying increased risk of COVID-19 in elderly people and people with MetS. Additionally, we discuss current progresses in treating COVID-19 with focuses on vaccine development and antivirals, reinforcing therapeutic approaches to surmount the pandemic.

2. Possible Molecular Mechanisms Underlying Increased Risk of COVID-19 Complications Associated with Aging and MetS

2.1. Aging and COVID-19

The majority of deaths caused by COVID-19 has been occurred in people over the age of 65. Age is thus clearly the most notable risk factor for COVID-19 mortality [20]. Similarly, older people are disproportionately affected by human coronaviruses and influenza viruses [21], yet except vaccines, no apparent therapeutic strategies have been suggested to protect this fraction of the population. A study in the UK (109,000 randomly selected teenagers and adults) showed that the infection fatality ratio (IFR) was close to zero (15–44-year-olds), whereas increasing to 3.1% (65–74-year-olds) and to 11.6% (older than 74 years) [22]. A comparable tendency was observed in another study from Spain (61,000 randomly selected residents); the overall IFR was close to zero (under 50 years old), whereas 11.6% for men and 4.6% for women (80 years old and over) [23]. Importantly, the age-specific COVID-19-associated death data (45 countries and 22 seroprevalence studies) indicated that spreading the virus in nursing homes or elderly-care facilities appears to be an essential factor [24].
Age-related changes of the immune system play critical roles in bestowing COVID-19 susceptibility: impair the body’s ability to carry away the SARS-CoV-2 virus and trigger cytokine storms, causing disseminated intervascular coagulation (DIC) and multi-organ injury as well as failure (Figure 1) [25]. The aging immune system undergoes immunosenescence, a progressive diminution in immune function [25]. The other representative immune system change during aging is inflammaging, a persisting elevation in systemic inflammation [4].
Within immunosenescence, there are: 1) innate immunosenescence, including inefficient recognition of pathogens and macrophage activation, and a decrease in natural killer (NK) cell cytotoxicity, and 2) adaptive immunosenescence, such as thymic degeneration and deposition of anergic memory lymphocytes [25]. Alveolar macrophages (AMs) are phagocytes that reside in the pulmonary alveoli. AMs display impaired function during aging; significant reduction of their conversion potential between pro- and anti-inflammatory phenotype [26]; downregulation of Toll-like receptor (TLR) expression and downstream signaling as well as pro-inflammatory and immunomodulatory cytokine production [8]. In older individuals, AMs exhibit diminished capacity to become pro-inflammatory state upon encountering pathogen, thus, spurring COVID-19 in its early stages, whereas AMs are correlated with the excessive lung damage in advanced stages of COVID-19 [25]. Moreover, severe COVID-19 patients had less expanded bronchoalveolar CD8+ T cells [27]; reduction and functional exhaustion of T cells were also observed in COVID-19 patients with high levels of the immune-exhaustion marker programmed cell death-1 (PD-1) in peripheral blood T cells [28].
Multiple evidence implicates that the cytokine storm, an uncontrollable inflammatory response by the immune system [29], is primarily derived from inflammaging, exacerbated by obesity and microbial dysbiosis [30,31]. In rodent animal models, higher risk of cytokine storm syndrome is closely correlated with inflammaging [32]; in humans, excessive basal circulating levels of pro-inflammatory cytokines, including interleukin (IL)-6, tumor necrosis factor (TNF)-α, IL-1α and C-reactive protein (CRP), are associated with age [33,34]. An age-associated decline in nicotinamide adenine dinucleotide (NAD+) results in de-repression of nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3), the major protein component of the inflammasome [35,36,37,38]. It was shown that SARS-CoV-2 proteins enhance the activity of poly-ADP-ribose polymerases (PARPs) PARP9, -10, -12, and -14, and decrease NAD+ levels [39]. Thus, it is likely that the decline of NAD+ levels, aggravated by COVID-19, increases the activity of NLRP3 and triggers cytokine storms in COVID-19 patients; restoring normal NAD+ levels likely alleviates COVID-19 symptoms.
Aging changes the glycome through non-enzymatic glycation; after exposure to sugars, proteins and lipids become glycated and develop advanced glycation end products (AGEs) [40]. AGEs form in hyperglycemic conditions and contribute to the pathology of many age-related, metabolic diseases and oxidative stress as well as inflammation [41]. The NLRP3 inflammasome activation in lipopolysaccharide-primed macrophages was shown to be induced by SARS-CoV 3a [42]; AGEs may escalate COVID-19 severity in elderly people by hindering NLRP3 inflammasome-mediated innate immune responses during the early stages of viral infection [43]. Furthermore, DIC seen in COVID-19 patients is likely to be triggered by AGEs considering their active role in pro-coagulation pathways [41].
Increased age is clearly the key risk factor for COVID-19 mortality [20]. Accordingly, as prophylactic approaches, not only vaccination but also activation of the body’s defenses against aging by using geroprotectors are in consideration. Low dose mechanistic target of rapamycin (mTOR) inhibitors was shown to reduce rates of infection and improved immunity and response to influenza vaccination in elderly [44,45]; consistently, metformin that inhibits the mTOR pathway and activates 5′-AMP-activated protein kinase (AMPK) as well as lowers blood glucose levels has been proposed to hamper severe SARS-CoV-2 infection in elderly [46].

2.2. Obesity and COVID-19

Multiple studies suggest that enhanced body mass index (BMI) is a critical indicator of adverse diagnosis of COVID-19 patients [47]. The data from Shenzhen, China demonstrated that obese and overweight patients were susceptible to acquire severe pneumonia compared with normal-weight patients [48]; a study in France described that obesity (BMI > 35 kg/m2) escalated the risk for ventilation [49]; another study from the United Kingdom, obesity was a risk factor for mortality with a significant BMI gradient [50]. These studies indicate that obesity is strongly correlated with severe COVID-19 and death.
Various pathological molecular linkages have been proposed for the detrimental prognosis in obese COVID-19 patients (Figure 2) [51]. Adipose tissue (AT) in individuals with morbid obesity is characterized by low-grade inflammatory state altering innate and adaptive immunity [52,53]. AT consists of mature adipocytes and several types of stromal cells, including fibroblasts, adipose-derived mesenchymal stromal/stem cells (ASCs), endothelial cells, and various immune cells [54]. Morbid obesity has been shown to 1) compromise functions and properties of ASCs, changing their role from a pivotal regulator in AT to a threatening foe that induces hypoxia and secrets pro-inflammatory cytokines, and 2) lead to the loss of functional mesenchymal stem/stromal cells (MSCs) in various organs, including lung and brain [55,56]. Obese patients also often have impaired pulmonary function with reduced lung volumes and diaphragmatic strength, intensified airway resistance, and blunted gas exchange [57]. Accordingly, COVID-19 patients with obesity likely undergo a more serious respiratory damage [58]. Inversely, obese ASCs/MSCs adversely influence the immune response and further increase systemic inflammation [55].
AT expresses several receptors and enzymes for SARS-CoV-2 entry into and exit from cells, including the receptors ACE2, dipeptidyl peptidase 4 (DPP4), and cluster of differentiation (CD147), and one of the priming proteases, furin, which are upregulated in obese patients [59,60,61,62,63,64,65,66,67]. Consequently, diseased adipose tissues could be attacked by SARS-CoV-2, increasing inflammation/immune response and causing multi-organ failure [60].
A fine tuning of proinflammatory/pro-coagulant and anti-inflammatory/anti-coagulant states of the endothelium is disturbed by obesity-associated chronic inflammatory process, resulting in vascular endothelial dysfunction [68]. Importantly, ACE2 receptor and the priming proteases, furin and transmembrane protease serine 2 (TMPRSS2), are highly expressed in vascular endothelium [69]. Additionally, obesity-related inflammation and metabolic dysregulation lead to changes in nitric oxide (NO) and reactive oxygen species (ROS). Endothelial nitric oxide synthase (eNOS)-generated NO relaxes vascular smooth muscle cells, increases blood flow, and represses the release of mediators that recruit proinflammatory cell populations to the endothelium [70,71]; NO produced by inducible nitric oxide synthase (iNOS) in adipocytes and proinflammatory macrophages is toxic levels and increased in obesity [72]; ROS are often substantially raised in obesity and gives rise to perilous pathological changes [73]. Accordingly, the endothelium of patients with obesity are likely to be highly vulnerable to SARS-CoV-2 infection.
Multiple obesity-related factors activate the proinflammatory immune cells, including dendritic cells (DCs), M1 macrophages, T helper type 1 (Th1) cells, and CD8+ T cells [51]. Hypernutrition and saturated free fatty acids (FFAs) stimulate expression of TLRs in DCs, especially TLR2 and TLR4 that activate M1 macrophages [74]; activation of M1 macrophages outside of AT are further induced by upregulated secretion of AT inflammatory cytokines [75]. Additionally, elevated levels of lipopolysaccharide and adenosine triphosphate activate the NLRP3 inflammasome [76]. The secretion of crucial adipocytokines also increases the production of TNF-α, IL-6, and IL-12, resulting in primarily pathogenic proinflammatory Th1 cell population [77]. Unsurprisingly, the severity and mortality rate of obese individuals were increased in multiple infectious diseases, such as influenza [18,78]. Even after vaccination, individuals with obesity had a higher risk of influenza or influenza-like illnesses compared to healthy lean individuals [79]; their influenza specific antibody titer was dramatically reduced one year after vaccination [80,81]. Accordingly, diminished immune response provoked by obesity should be taken into account for developing SARS-CoV-2 vaccines as a key prophylactic approach.
Importantly, one of the key hallmarks of obesity is adaptations in cardiac structure and function associated with a highly increased risk of cardiovascular diseases (CVD) [82]. Based on the previous coronavirus and influenza epidemics, pre-existing CVD and cardiovascular (CV) risk factors likely enhance vulnerability to COVID-19 [83,84,85,86,87,88,89,90]. Further, COVID-19 can worsen underlying CVD and cause de novo cardiac complications [91]. Several mechanisms are responsible for cardiovascular complications in COVID-19 [91,92,93]. SARS-CoV-2 can directly damage cardiomyocytes by binding to ACE2 and altering ACE2 signaling pathways with subsequent acute myocardial injury [92,93]. SARS-CoV-2 spreads through respiratory mucosa and concomitantly infect other cells, resulting in systemic inflammatory response and cytokine storm [94]. The systemic infection coupled with hypoxia can impair myocardial oxygen demand-supply relationship and lead to acute myocardial injury [91]. Additionally, systemic inflammation after infection can give rise to reduction in coronary blood flow (referred as acute coronary syndromes (ACS)), decrease in oxygen supply, destabilization of coronary plaque, and microthrombogenesis, which result in acute myocardial infarction [94,95,96]. Adverse effects of various therapies for COVID-19 and electrolyte imbalances can also have deleterious effects on the CV system [91]. In the case study of 187 patients with COVID-19, patients with underlying CVD, including hypertension, coronary heart disease, and cardiomyopathy, were more prone to experience myocardial injury during the course of COVID-19 [94]. Patients with underlying CVD with myocardial injury (demonstrated by escalation of troponin T (TnT) levels) had the highest mortality with the shortest survival term [94]. Interestingly, while patients with myocardial injury showed markedly increased mortality rate, patients with underlying CVD but with normal TnT levels experienced a more favorable prognosis [94]. These data suggest that myocardial biomarkers should be assessed in patients with CVD who develop COVID-19 for risk categorization and intervention. In the cohort study of 416 patients with confirmed COVID-19, patients with cardiac injury had higher mortality than those without cardiac injury; these patients had a history of coronary heart disease and hypertension, indicating that preexisting CV diseases were more susceptible COVID-19-induced heart injury [97]. Thus, the presence of pre-existing CV disease and/or development of acute cardiac injury are closely related to dramatically worse outcomes in COVID-19 patients.

2.3. Diabetes and COVID-19

The World Health Organization (WHO) estimates 422 million individuals in the world have diabetes with its prevalence rising rapidly; up to 95% of them suffer from T2DM. In the United States (US), 10.5% of the US population have been diagnosed with diabetes and 34.5% of the adult US population have prediabetes with the risk of onset of T2DM (National Diabetes Statistics Report, 2020). It is the unprecedented aging and obesity that are major contributors to the epidemic of T2DM. T2DM advances from prediabetes with impaired glucose tolerance (IGT) and/or MetS to overt diabetes with a diminution in functional β-cell mass [98,99]. Clinical studies have shown that decline in β-cell function derived from multiple risk factors, including obesity, inflammation, and IR, starts several years before the diagnosis of T2DM; fasting plasma glucose levels gradually increase for 10-12 years before diagnosis of T2DM [100]. At the time of the clinical onset of T2DM, patients have significantly fewer β-cells than healthy individuals as β-cells are metabolically afflicted and prone to apoptosis during the period of continual hyperglycemia [100].
T2DM is a pivotal predictor for COVID-19 severity and its complications [47]. Current data suggest worse outcomes in patients with preexisting diabetes [47]. A study in Hubei Province, China (a cohort of confirmed COVID-19 cases) showed that the fatality rate was higher in patients with T2DM compared to nondiabetic individuals (7.8% vs. 2.7%) [101]. Hyperglycemia is likely to modulate the risk of complications; the risk of death was lower in the subgroup with blood glucose (<7.5 (5.2–7.5) mmol/L; 1.1%) compared with poorly controlled blood glucose group (>7.6 (7.6–14.3) mmol/L; 11.0%) [101]. Similarly, analysis of the health data in the United Kingdom (17.4 million adults) revealed that uncontrolled diabetes was an independent risk for death from COVID-19 [50]. A multicenter observational study in France (diabetic patients hospitalized for COVID-19) also demonstrated that BMI was independently correlated with mechanical ventilation and/or death [102]. These studies suggest the connection between diabetes and COVID-19 mortality/complications.
Multiple mechanisms play roles in increasing the susceptibility of complications in diabetic COVID-19 patients [103]. Basically, as aforementioned, diabetes is tightly associated with the hallmarks and progression of obesity. Circulating levels of furin are elevated in patients with diabetes [104]. As observed in rodent models of diabetes [105,106], enhanced ACE2 expression in diverse tissues likely favors increased cellular binding and infection of SARS-CoV-2 [107]. Administration of insulin in diabetic mice decreased ACE2 protein expression in the lungs [105,106]. ACE2 is a part of the renin-angiotensin system (RAS) controlling blood pressure, balance of fluid and electrolyte, and systemic vascular resistance [25]. ACE catalyzes the conversion of the prohormone, angiotensin (Ang) I to the octapeptide, AngII, inducing vasoconstriction and proliferation; ACE2 converts AngII to Ang-(1–7), stimulating vasodilatation and suppressing cell growth [103]. Accordingly, suppressing ACE2 could hinder viral entry but may induce vasoconstriction and hypertension. Increased ratio of pulmonary ACE/ACE2 activity, which favors AngII generation, was observed in patients with ARDS [108]. Upon binding to ACE2, SARS-CoV was shown to downregulate ACE2 expression, leading to acute pulmonary damage via AngII [109]. Thus, in the setting of low ACE2 expression, upregulating ACE2 using agents that induce hypoglycemia and treat hypertension can be beneficial [103]. For an ACE2-targeted therapeutic strategy to prevent viral entry, infusion of human recombinant soluble ACE2 into the airway or bloodstream can be promising [110].
Altered immune response in diabetic patients is closely associated with a chronic low-level inflammation, lowered activity of NK cells, increased T cell activation, elevated inflammatory cytokine profile, and decreased Treg function, leading to dysregulated immune function in COVID-19 patients [55,111,112]. Microvascular endothelial dysfunction correlated with several key factors, including chronic inflammation, increased expression of iNOS, and elevated production of ROS, is also a frequent manifestation in diabetic patients [113]. Accordingly, dysfunctional shift of endothelial cells toward pro-thrombotic and pro-atherogenic states instigates severe impairment of COVID-19 on the cardiovascular system [51]. Diabetes, like obesity, is characterized by hypercoagulable state as well [114], featuring hypofibrinolysis and elevated levels of plasminogen activator inhibitor-1 (PAI-1) complement as well as increased platelet aggregation [115,116]. In patients with established T2DM, neutrophil extracellular traps (NETs), complexes of chromosomal DNA, histones, and granule proteins, were higher compared with healthy individuals [117,118]. Severely ill COVID-19 patients display coagulopathy/thrombosis [119], which is worsened by acute hyperglycemia in patients with diabetes [112]. Thus, optimal management of hyperglycemia is crucial for COVID-19 patients with diabetes [47,120].

3. Current Progresses in Prophylactic and Therapeutic Treatment Options for COVID-19

3.1. Vaccination

Mass prophylactic vaccination is likely the only viable path to cost-effectively curb the COVID-19 pandemic. Following are some of the vaccine platforms that are being utilized for COVID-19 vaccine development in clinical trials and licensed in use on the market (Table 1) [121,122].

3.1.1. Inactivated Vaccines

The virulent antigen or infectious viruses killed through physical or chemical processes are the basis of inactivated vaccines [121]. In case of SARS-CoV-2, the viruses are cultured in Vero cells and chemically inactivated [121]. Successful inactivated human pathogens as vaccines include Influenza, Hepatitis A and poliomyelitis [123,124,125]. The inactivated vaccines are safer to use and relatively easy to produce [126]; they express surface antigens that retain their epitope conformations to induce strong preventative humoral responses [127]. However, the reduced production of SARS-CoV-2 virus in cell culture and the requirement of biosafety level 3 (BSL3) facilities can lead to limited yield. The inactivated vaccines are commonly administered intramuscularly and adjuvanted with alum (aluminium hydroxide) [122]. Several inactivated vaccines against SARS-CoV-2 are currently being produced and some have entered in the clinical trials (Table 1). Examples of inactivated vaccine candidates include CoronaVac (Sinovac, Beijing, China) [128], BBIBP-CorV (Sinopharm, Shanghai, China) [129], Covaxin (Bharat Biotech, Hyderabad, India), and QazCovid-In (Research Institute for Biological Safety Problems, Gvardeysk, Kazakhstan), which are in clinical Phase III trials (Table 1).

3.1.2. Live Attenuated Vaccines

Live attenuated vaccines use the live but a weakened version of the living virus [130], eliciting similar immune responses to natural infection without disease [131]. Generally, the viruses are exposed to unfavorable conditions (e.g., growth at lower temperature or in non-human cells) or genetically manipulated (e.g., modifying the viral genes through mutagenesis) [132,133,134,135,136]. Live attenuated seasonal influenza vaccine is an adequate example [137,138,139]. Important advantages of these vaccines are that they can be given intranasally, protecting the upper respiratory tract, which is the major portal of entry of SARS-CoV-2 [140,141,142]; they can induce both humoral and cellular immune responses. However, due to the live nature of the virus, this vaccine platform is less safe. Moreover, SARS-CoV-2 is novel coronavirus with a large (approximately 30,000 nucleotides) RNA, thus, it is challenging to generate genetically modified, live attenuated vaccine candidates [143]. Accordingly, only four live-attenuated vaccines are in preclinical trials; the one developed by Codagenix in collaboration with the Serum Institute of India is currently in clinical phase I. Intriguingly, “SARS-CoV-2/human/Korea/CNUHV03/2020” vaccine was generated by growing SARS-CoV-2 from 37 °C to 22 °C in Vero cells [144]. A single intranasal administration of this vaccine induced high titers of neutralizing antibodies and mucosal IgA antibodies as well as cellular immune responses in K18-hACE2 mice without detrimental systemic effects [144].

3.1.3. Recombinant Protein Vaccines

Recombinant protein vaccines for SARS-CoV-2 can be classified into recombinant spike-protein-based vaccines, recombinant receptor binding domain (RBD)-based-vaccines, and virus-like particle (VLP)-based-vaccines, which carry no genome but display the spike ‘S’ protein on their surface [121,122]. A variety of eukaryotic expression systems are utilized to manufacture these recombinant proteins; even prokaryotic E. coli is used to express RBD [145,146,147]. These vaccines can be produced without handling live virus; considerable expertise has been acquired to produce recombinant subunit vaccines as shown in licensed FluBlok vaccine for influenza [148,149,150]. However, the spike ‘S’ protein is relatively hard to express, raising the subsequent questions of low yield and number of doses. While the RBD peptide may be easy to produce [151], the absence of other neutralizing epitopes present on the full-length S protein likely leads to the antigenic drift in RBD-based vaccines. Many recombinant protein vaccines are currently being produced and evaluated, including NVX-CoV2373 (Novavax, Gaithersburg, MD, USA; recombinant full-length S protein nanoparticle) [152] and ZF2001 (Anhui Zhifei Longcom Biopharmaceutical, Chongqing, China; RBD-dimer), which are in clinical Phase III trials (Table 1).

3.1.4. Replication-Incompetent/-Competent Vector Vaccines

Delivery vectors, such as viral vectors, are utilized to develop expression vector-based vaccines. Specifically, for viral vector-based vaccines, replication-incompetent or -competent viral backbone is engineered to express the target-pathogen-derived antigens.
The majority of the replication-incompetent vectors are based on recombinant adenovirus (AdV) vectors [153]; it is not necessary to handle live virus; considerable expertise has been acquired to produce large quantities in manufacturing the recombinant adenovirus vaccines; both B- and T-cell responses are induced by the vectors [121]. While pre-existing vector immunity affect some of these vectors, using adequate vector types, including adeno-associated viruses that do not induce much immunity by themselves, can overcome this complication [154]. Several replication-incompetent vector vaccines are in use, such as Ad26.COV2.S (Janssen Pharmaceutical Companies of Johnson & Johnson, Titusville, NJ, USA; authorized) [155], ChAdOx1 nCoV-19 (AstraZeneca, Cambridge, UK/University of Oxford, Oxford, UK; authorized) [156,157], Sputnik V (Gameleya Research Institute, Moscow, Russia; clinical phase III) [158], and Ad5-nCoV (CanSino Biologics, Tianjin, China; clinical phase III) [153,154] (Table 1).
Replication-competent vectors are derived from attenuated viruses; to reduce the pathogenicity, the genes that are nonessential for viral replication in cultured cells in vitro are modified [159]. This approach induces more strong immunity owing to the vector’s capability to propagate in the vaccinated individuals [121]. Examples of replication-competent vector vaccines include DelNS1-2019-nCoV-RBD-OPT1 (Beijing Wantai Biological, Beijing, China; clinical phase II) and V591 (Institut Pasteur, Paris, France/Themis, Kenilworth, NJ, USA; clinical phase I/II).

3.1.5. DNA Vaccines

COVID-19 DNA vaccines are plasmid DNA encoding the SARS-CoV-2 spike ‘S’ gene under a mammalian promoter [121,122]. They can be produced at large scale in bacteria and offer high stability, whereas they often show low immunogenicity. Accordingly, booster doses and intracellular delivery devices, such as electroporators, are needed to attain the expected effect in vivo. There are several SARS-CoV-2 DNA vaccines currently in clinical trial stages, such as ZyCov-D (Zydus Cadila, Ahmedabad, India; clinical phase III), INO-4800 (Inovio Pharmaceuticals, Plymouth Meeting, PA, USA; clinical phase II/III) [160], and AG0301&AG0302 (AnGes, Osaka, Japan/Osaka University, Osaka, Japan/Takara Bio, Shiga, Japan; clinical phase II/III) (Table 1).

3.1.6. RNA Vaccines

Two major types of RNA vaccines have been used against infectious pathogen: non-amplifying mRNA vaccines, either conventional or chemically modified base incorporated, and self-amplifying (or replicon) RNA vaccines retaining auto-replicative activity derived from a RNA virus vector [161]. Due to the self-replicative properties, self-amplifying RNA vaccines require a lower dose of RNA than non-amplifying mRNA vaccines [162]. RNA vaccines have a key advantage compared to DNA vaccines that require an additional transcription step in in vivo systems; RNA vaccines can be easily synthesized in vitro in the lab. Furthermore, RNA vaccines reduce the risk of insertional mutagenesis. However, RNA vaccines require frozen storage due to their thermolabile nature (unstable at high temperatures). Consequently, modifications can be introduced for their safety, stability, and immunogenicity [163]. Lipid nanoparticle (LNP) platform has been utilized for RNA vaccine delivery [164,165]. There are several SARS-CoV-2 RNA-vaccines in clinical trials or licensed for vaccination on the market, including BNT162b2 (Pfizer, NY, NY, USA/BioNTech, Mainz, Germany; authorized) [166], mRNA-1273 (Moderna, Cambridge, MA, USA/NIAID, Rockville, MD, USA; authorized) [167], CVnCoV (CureVac, Tübingen, Germany; clinical phase III) (Table 1). Specifically, Pfizer’s BNT162b2 RNA vaccine is a LNP-formulated, nucleoside-modified (two proline mutations) RNA vaccine that encodes a prefusion stabilized, membrane-anchored SARS-CoV-2 full-length spike protein [166]. A report from a clinical trial (ClinicalTrials.gov number, NCT04368728; accessed 04/10/2021) demonstrated that BNT162b2 was 95% effective in preventing Covid-19; over 90% vaccine efficacy was observed even in the presence of coexisting conditions (e.g., obesity) [166]. mRNA-1273 from Moderna encodes the full-length spike ‘S’ protein with two stabilizing mutations and is encapsulated in LNPs for delivery. Phase III clinical trials (ClinicalTrials.gov number, NCT04470427; accessed 04/10/2021) with geriatric patients demonstrated that mRNA-1273 showed 95% efficacy across all demographics [168].

3.2. Antivirals and Therapeutics

There are three general methods to search for the promising antivirals and therapeutic drugs of human SARS-CoV-2: utilization of the current broad-spectrum drugs, testing chemical libraries with existing compounds or databases, and development of new specific drugs based on the genome and biophysical understanding of SARS-CoV-2 [169]. Following are some of the antivirals and therapeutics for treating COVID-19 in clinical trials and in use (Table 2) [170,171].

3.2.1. Antivirals

Remdesivir

Remdesivir (RDV) is one of the leading antivirals against SARS-CoV-2. While it was developed for treating Ebola virus [172], it displays the activity against other RNA viruses, such as paramyxoviruses, pneumoviruses, and coronaviruses (e.g., SARS-CoV and MERS-CoV) [173]. RDV, also known as GS-5734, is a 1′-cyano-substituted adenosine nucleotide analog [174]. As a prodrug, RDV is metabolized in cells and tissues to an active nucleoside triphosphate (RDV-TP; GS-443902) that inhibits viral RNA-dependent RNA polymerases (RdRps) by incorporating RDV-TP into nascent RNA chains [175,176]. The Adaptive COVID-19 Treatment Trial (ACTT; NCT04280705) showed that RDV was more efficient than a placebo in treating hospitalized COVID-19 patients [177]. RDV treatment shortened the time to recovery (11 days versus 15 days) and reduced respiratory infection as well as mortality compared to placebo [177]. RDV was approved by the US Food and Drug Administration (FDA) for treating adults and pediatric patients requiring hospitalization for COVID-19; it is administered via intravenous (IV) infusion [171]. There are several ongoing clinical trials with RDV, including inhaled RDV formulation for early-stage COVID-19 [170].

Favipiravir

Favipiravir is an antiviral approved for treating influenza in Japan; it exhibits antiviral activity against several other viruses, including chikungunya virus, Rift Valley fever virus, hepatitis C virus (HCV), and the West Nile virus by inhibiting RdRp [178,179,180,181,182]. In an open-label control study in China, favipiravir treatment showed better therapeutic responses on COVID-19 with regard to disease progression and SARS-CoV-2 clearance [183]; other trials have reported that favipiravir alleviated some of the COVID-19 symptoms, such as cough, and stimulated defervescence [184]. These studies suggest the potential beneficial effects of favipiravir on patients with mild COVID-19. There are various ongoing clinical trials with favipiravir; it has been approved for use in multiple countries.

Lopinavir

Lopinavir (LPV) is a human immunodeficiency virus (HIV)-1 protease inhibitor administered in fixed-dose combination with ritonavir (LPV/r). Most of the protease inhibitory effects are attributed to LPV, while ritonavir enhances the pharmacokinetic and pharmacodynamic properties of LPV [185]. SARS-CoV, MERS CoV, and SARS-CoV-2 coronaviruses are RNA viruses like HIV; the nonstructural protein of coronaviruses, especially the main protease 3C-like protease (3CLpro), is involved in the proteolysis of the replicase polyprotein, which is essential for viral infection [186]. As shown in treating patients in South Korea and in China in early SARS-CoV-2 outbreak, LPV/r was expected to be beneficial for COVID-19 treatment based on the sequence similarity of the CoV proteases [187,188]. However, the results of the Efficacy of Lopinavir Plus Ritonavir and Arbidol Against Novel Coronavirus Infection (ELACOI) and Randomized Evaluation of COVID-19 Therapy (RECOVERY) as well as WHO’s Solidarity trial showed little or no benefit of LPV/r to hospitalized COVID-19 patients [189,190,191]. Several clinical trials are currently exploring the effects of LPV/r as prophylaxis or for early-stage COVID-19 [170].

Ribavirin

Ribavirin is a guanosine analog that interferes with viral RNA synthesis by inhibiting viral RdRp and mRNA capping [192]. In primate models, using rhesus macaques, ribavirin alone or in combination with interferon (IFN)-α2b reduced virus replication of MERS-CoV and ameliorated the clinical outcomes with moderation of the host response [193]. Consistent with the positive clinical outcomes in some MERS cases using the combination of ribavirin with LPV/r and IFNs [194,195], the combination of ribavirin with IFN-β1b and LPV/r alleviated symptoms in patients with mild to moderate COVID-19 [196]. Contrarily, combinations of ribavirin + IFN-α, LPV/r + IFN-α, and ribavirin + LPV/r + IFN-α, or combination of ribavirin + LPV/r showed no significantly different antiviral effects or even adverse effects in a randomized open-labeled prospective study with mild to moderate COVID-19 patients [197]. Ongoing clinical trials are underway to determine the potential of ribavirin in COVID-19 treatment [170].

Umifenovir

Umifenovir (brand name Arbidol) is an antiviral approved in China and Russia for treating influenza. It binds in a hydrophobic cavity in the hemagglutinin (HA) trimer stem, inhibiting viral membrane fusion in the low pH of the endosome [198]. A retrospective study suggested that umifenovir treatment increased the discharging rate with a decrease in the mortality rate of COVID-19 patients [199]; the results of a randomized, controlled trial demonstrated that treatment of hydroxychloroquine (HCQ) + umifenovir resulted in substantially shorter duration of hospitalization and decreased intensive care unit (ICU) admission rates than HCQ + LPV/r treatment in hospitalized COVID-19 patients [200]. However, another retrospective study demonstrated little effect of umifenovir on improving clinical consequences in non-ICU patients [201], suggesting more trials for accessing the effects of umifenovir in COVID-19.

Chloroquine and Hydroxychloroquine

Chloroquine (CQ) and hydroxychloroquine (HCQ) are used to prevent/treat malaria. CQ has been shown to inhibit the replication of different CoVs, including SARS-CoV, MERS-CoV, and SARS-CoV-2 among others; HCQ was shown to have potent in vitro inhibitory effects against SARS-CoV-2 replication [202]. CQ and HCQ increase endosomal pH, thus inhibiting virus-endosome fusion for viral endocytosis [203,204]. An open-label non-randomized clinical trial for HCQ with azithromycin, which was shown to be active in vitro against Zika and Ebola viruses and prevent severe respiratory tract infections [205,206,207], demonstrated that HCQ significantly reduced viral load in COVID-19 patients; its effect was reinforced by azithromycin [208]. However, the final results of the RECOVERY groups HCQ trial showed that hospitalized COVID-19 patients receiving HCQ showed similar mortality rate to the patients without treatment [209]. Several trials are still ongoing to determine the effects of CQ and HCQ as prophylaxis or for early-stage COVID-19 [170].

3.2.2. Immune-Based Therapeutics

Neutralizing Antibodies

Convalescent plasma (CP) from individuals following resolution of infection contains neutralizing Abs (NAbs) that bind to the pathogen and neutralize its biological effect [210]. Accordingly, plasma exchange can be a favorable option for treating COVID-19 patients. While a clinical trial from eastern Anatolia showed that patients with early-stage COVID-19 who did not need mechanical ventilation improved with CP treatment [211], the largest completed CP study to date (PlasmAr; ClinicalTrials.gov number, NCT04383535; accessed 04/10/2021) indicated no significant differences in clinical status or overall mortality between COVID-19 patients with severe pneumonia treated with CP and those who received placebo [212]. Furthermore, considering the potential risks arising from CP therapy, including viral or bacterial infections, lung damage, and allergic reactions [213], the use of mAbs and mAb cocktails have been presented. Several studies demonstrated NAbs that can be used for COVID-19 treatment by targeting diverse epitopes on the spike ‘S’ protein [214,215,216,217]. Administration of LY-CoV555 (bamlanivimab; granted emergency use authorization by the US-FDA), an IgG1targeting the SARS-CoV-2 ‘S’ protein, into the patients diagnosed mild or moderate CoVID-19 resulted in accelerating the natural decline in viral load over time [218]. The REGN-COV2 (granted emergency use authorization by the US-FDA), an antibody cocktail of two anti-SARS-CoV-2 ‘S’ protein Abs (REGN10933 + REGN10987), diminished viral load [219].

Cytokine Inhibitors: Tocilizumab

The primary cause of death in seriously ill COVID-19 patients is ARDS and multi-organ system failure [171]. The heightened synthesis of inflammatory cytokines/chemokines leading to cytokine release syndrome (CRS) has been implicated as a main culprit; IL-6 is a key inflammatory driver, thus, can be a biomarker indicating worsening of COVID-19 [220,221]. Tocilizumab is a recombinant humanized monoclonal antibody that competitively inhibits the binding of IL-6 to soluble and membrane-bound IL-6 receptor (IL-6R) [222]. It has been approved by the FDA mainly for treating rheumatoid arthritis (RA) [222]. Several studies demonstrated that tocilizumab therapy reduced ICU admissions and mortality in critical COVID-19 patients; treatment with low-dose tocilizumab in non-critical hospitalized COVID-19 patients alleviated inflammation with faster defervescence [223,224,225,226,227,228]. However, contradicting results with no reduction in mortality and progression to mechanical ventilation were observed in trials for tocilizumab [229,230]. These contradictory results underscore that larger placebo-controlled studies are required to determine the effects of tocilizumab in COVID-19.

JAK Inhibitors: Baricitinib

Four mammalian Janus kinases (JAKs), JAK1, JAK2, JAK3, and tyrosine kinase 2 (Tyk2) are protein tyrosine kinases [231]. The JAK/signal transducer and activator of transcription (STAT) pathway plays important roles in cytokine-/interferon-induced inflammatory responses, thus, JAK inhibition can be a potential resource for treating COVID-19 [232]. Baricitinib is a JAK1/2 inhibitor licensed for the treatment of RA [233]; it has anti-viral effects by its affinity for adaptor-associated protein kinase 1 (AAK1), an adaptor protein-2 complex (AP2)-associated protein, thus, reducing SARS-CoV-2 endocytosis [234]. Baricitinib-therapy started in the early phase of COVID-19 reduced the fatality rate and ICU admission rate with decreased SARS-CoV-2 viral burden without serious adverse events [235,236]. A double-blind, randomized, placebo-controlled trial for baricitinib plus RDV reported that the combination reduced recovery time and accelerated improvement in clinical status of hospitalized COVID-19 patients, notably those under high-flow oxygen or noninvasive ventilation support [237]. Several trials to evaluate the benefits of baricitinib in treating moderate and severe COVID-19 are underway [170].

3.2.3. Statin Therapy

Statins reduce the synthesis of cholesterol in the liver by competitively repressing the hydroxymethyl-glutaryl coenzyme A (HMG-CoA) reductase [238]. Therefore, statins have been widely used to prevent and treat CVD as obesity-induced dyslipidemia is one of traditional CV risk factors [239]. Statins have also demonstrated inhibitory effects on several viruses, including influenza [240]. The role of statins in treating COVID-19 has been proposed as their cholesterol-lowering capacity can destabilize the formation of lipid rafts that are needed for ACE2-mediated SARS-CoV-2 infection [241,242]. Moreover, upon entering the cell, SARS-CoV-2 downregulates expression of ACE2, thus reducing its protective effects on various tissues and leading to organ injury; statins have been shown to increase the expression of ACE2 in animal models [243,244,245,246,247]. Statins also have pleiotropic effects: restoring vascular redox balance, inhibiting the thrombogenic response, and decreasing oxidative stress and inflammation by inhibiting the MYD88-NF-κB proinflammatory pathway [243,248,249]. Intriguingly, increased secretion of anti-inflammatory cytokines, such as IL-2 and IL-10, seems to represent a distinctive feature of COVID-19 [95]. While the dysregulation of secreted pro- and anti-inflammatory cytokines likely plays a critical role in the progression of ARDS and the severity of the diagnosis, cholesterol biosynthesis pathway was shown to be essential for dictating the CD4+ T cell switch from the effector (Th1 cell) to the anti-inflammatory (IL-10-secreting Th2 cell) phenotype [249,250]. A retrospective study on 13,981 patients with COVID-19 in Hubei Province, China reported that in-hospital use of statins compared to non-statin use was significantly associated with a lower risk of death and a less inflammatory response during the entire hospitalization period [251]. Another retrospective single-center study (University of California San Diego Health) showed that statin use during the 30 days prior to admission for COVID-19 was associated with a lower risk of developing severe COVID-19, and a faster time to recovery among patients without severe disease [252]. However, an observational multicenter study with 842 COVID-19 patients showed that statin therapy was associated with a worse disease severity (but nevertheless, not with in-hospital mortality) [253]. Thus, statin use can rather be considered as a proxy of underlying comorbidities; in patients on high-intensity statin treatment, the enhanced severity of underlying CV risk factors and CVD can explain the higher severity of COVID-19 [253]. Accordingly, further prospective studies and randomized controlled clinical trials are needed to more definitely determine the overall clinical benefits of statin treatment for COVID-19-related pathologies.

3.2.4. Anticoagulation Therapy

Many severe COVID-19 patients show coagulation abnormalities with an increased risk of death, indicating the presence of a hypercoagulable state also detected in obesity and diabetes [114,254]. The coagulopathy associated with COVID-19 is a combination of low-grade DIC (mixture of thrombocytopenia, prolonged prothrombin time, and increased D-dimer) and confined pulmonary thrombotic microangiopathy, which can result in organ dysfunction in the most severely affected patients [254]. Several distinctive pulmonary vascular pathological features of COVID-19 are 1) severe endothelial injury associated with disrupted endothelial cell membranes, 2) widespread vascular thrombosis with microangiopathy, 3) occlusion of alveolar capillaries, and 4) substantial new vessel growth via intussusceptive angiogenesis [255,256]. Severe COVID-19 is also associated with increased concentrations of proinflammatory cytokines [95]. Specifically, IL-6 can induce tissue factor expression on mononuclear cells, which initiates coagulation activation and thrombin generation; TNF-α and IL-1 mediate a suppression of endogenous anticoagulant pathways [254]. A retrospective study in China (including 449 patients admitted to hospital with severe COVID-19 infection) showed a lower mortality in patients with COVID-19-associated coagulopathy who received prophylactic heparin than in patients not receiving anticoagulant treatment [257]. Particularly, in patients with increased concentrations of d-dimer (6 times the upper limit of normal), mortality was lower in heparin-treated patients than those not treated with heparin, while a prospective randomized controlled trial is needed to confirm these results [257]. Another study with 844 COVID-19 patients reported that oral anticoagulants (OACs) appeared to be ineffective in reducing mortality rate, whereas heparin resulted to be an effective treatment when lung disease was severe; larger studies are needed to confirm these findings [258]. Heparin inhibits cellular invasion by SARS-CoV-2 by binding to the spike S1 protein receptor binding domain and inducing a conformational change; it also exhibits anti-inflammatory effects, inhibiting inflammatory cell infiltration and dampening pro-inflammatory signals; these features may be critical in determining different outcomes compared to OACs [259,260]. Hence, the benefits of heparin treatment should be continuously evaluated, while considering the risk factors, including the increased likelihood of bleeding and other contraindications for anticoagulation.

4. Conclusions

SARS-CoV-2 caused a grievous pandemic of COVID-19. Accordingly, multiple clinical trials are in progress, evaluating treatment options for COVID-19, some of which have been listed in this review (Table 1 and Table 2). Vaccination could be the most effective prophylactic measure to prevent the ongoing COVID-19 pandemic as seen in continuing global vaccination programs with licensed COVID-19 vaccines (Table 1). Antivirals, including RDV and favipiravir, and NAbs, such as LY-CoV555 and REGN-COV2, have been successfully applied to treat the patients with COVID-19 whereas more trials are needed to adequately evaluate their effects. The severity of COVID-19 is strongly associated with age; there is an increased prevalence of MetS, such as obesity and diabetes, in people hospitalized with severe COVID-19 illness. Notably, the mRNA-1273 and BNT162b2 vaccines were equally protective in elderly people and individuals with obesity/T2DM [166,168]. Nevertheless, the long-term durability of immunogenicity has not been assessed; both the occurrence of adverse events after the second dose and more apprehensive information on the duration of protective immunity remain to be determined. Furthermore, comprehensive understanding of the underlying mechanisms of the adverse outcomes of COVID-19 illness in elderly people and hospitalized people with MetS will give insight into the development of disease-specific therapeutic interventions.

5. Future Research Directions

5.1. Therapeutic Perspectives

Data from retrospective observations indicate that glycemia optimization treatment likely reduces the risk of severe COVID-19 [261]. While GLP-1 receptor (GLP-1R) agonists are approved for weight loss, scant information is obtainable whether this class of drugs is reliable and beneficial in SARS-CoV-2-infected people with obesity, needless to say the necessity of the safety profiles of other glucose-lowering agents in people with clinical symptoms of COVID-19. Pre-clinical studies demonstrate that treatment of GLP-1R agonists attenuated virus-triggered pulmonary inflammation in mice [262,263,264], whereas data of how GLP-1R agonists function in experimental or clinical SARS-CoV-2 infection are not yet available. In addition, whether the risk of severe COVID-19 infection can be dampened by weight loss has not been systemically studied. Intriguingly, treatment of obese mice with metformin, not weight loss, was able to improve survival to influenza in obesity, suggesting an importance of pharmaceutical interventions [265].
Individuals with obesity and/or obesity-derived T2DM with prolonged immobility in the hospital are vulnerable for thromboembolic disease [261]. As shown in severely ill, hospitalized people with COVID-19 in Wuhan, China [266], one of the common pathophysiology for greater COVID-19 vulnerability in people with obesity and/or T2DM is their predisposition for developing coagulation-related complications. Consequently, the forthcoming results of various anti-coagulant strategies can be critical therapeutic options for preventing SARS-CoV-2-related coagulopathy.

5.2. Mechanistic Perspectives

The details of how much inflammation occurs and gene expression is dysregulated in metabolically important tissues are not readily available in active SARS-CoV-2 infection. While inflammatory responses in lung, brain, heart, kidney, and vascular tissues have been an intense focus of autopsy analyses [267], studies of the adipose tissues, pancreas, or endocrine organs are very limited. Furthermore, little molecular data is at hand from tissue biopsies of SARS-CoV-2-infected people with obesity and diabetes. Major conclusions regarding the mechanisms of viral entry and the features of glucose-lowering drugs are stemmed from preclinical studies or people without COVID-19 [268], thus, may be irrelevant for understanding human SARS-CoV-2 infection.
SARS-CoV-2 is incapable to propagate in wild-type mice owing to incompatibility of the spike ‘S’ protein with mouse ACE2 [269]. Thus, transgenic mice expressing human ACE2 in proper lung epithelial cells were established to model the replication and susceptibility of SARS-CoV-2 in vivo [270,271,272]. Recently, mouse-adapted SARS-CoV-2 (SARS-CoV-2 MA) was generated [273,274]. The SARS-CoV-2 MA model exhibited more clinically applicable phenotypes than those seen in HFH4-hACE2 transgenic mice expressing human ACE2 under the control of a lung ciliated epithelial cell-specific HFH4/FOXJ1 promoter [273]. Consequently, SARS-CoV-2 MA can be applied to the existing mouse models to better understand the impact of COVID-19 on obesity, such as genetically obese mouse models, including leptin-deficient (ob/ob) and leptin-receptor-deficient (db/db), as well as high-fat diet-induced-obese (DIO) mouse model [275,276,277]. Significant scientific gaps exist in the roles and mechanisms of host genetics and aging/MetS governing SARS-CoV-2 pathogenesis. Moreover, the detailed protective or pathogenic immune responses remain elusive, relating to COVID-19 severity. It is therefore of paramount importance to evaluate the performance of vaccines and therapeutics in the most vulnerable populations of SARS-CoV-2 infection.

Author Contributions

Conceptualization, C.-H.S. and S.-M.K.; writing—original draft preparation, C.-H.S.; writing—review and editing, C.-H.S., K.-H.K., S.J., S.-M.K.; visualization, C.-H.S.; supervision, project administration, and funding acquisition, S.-M.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Institutes of Health, AI093772.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

COVID-19Coronavirus disease 2019
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
T2DM Type 2 diabetes mellitus
ACE2 Angiotensin-converting enzyme 2
ARDS Acute respiratory distress syndrome
DM Diabetes mellitus
SASPSenescence-associated secretory phenotype
NKNatural killer
MetS Metabolic syndrome
IRInsulin resistance
HDLHigh-density lipoprotein
IFRInfection fatality ratio
DICDisseminated intervascular coagulation
AMsAlveolar macrophages
TLRToll-like receptor
PD-1Programmed cell death-1
ILInterleukin
TNFTumor necrosis factor
CRPC-reactive protein
NADNicotinamide adenine dinucleotide
NLRP3Nucleotide-binding domain (NOD)-like receptor protein 3
PARPsPoly-ADP-ribose polymerases
AGEs Advanced glycation end products
AMPK5′-AMP-activated protein kinase
BMIBody mass index
ATAdipose tissue
ASCsAdipose-derived mesenchymal stromal/stem cells
MSCsMesenchymal stem/stromal cells
DPP4Dipeptidyl peptidase 4
CD147Cluster of differentiation
TMPRSS2Transmembrane protease serine 2
NO Nitric oxide
ROS Reactive oxygen species
eNOS Endothelial nitric oxide synthase
iNOS Inducible nitric oxide synthase
DCsDendritic cells
Th1T helper type 1
FFAsFree fatty acids
CVD Cardiovascular diseases
CV Cardiovascular
ACSAcute coronary syndromes
WHOWorld Health Organization
USUnited States
IGTImpaired glucose tolerance
RAS Renin-angiotensin system
Ang Angiotensin
GLP-1 Glucagon-like peptide-1
TZDs Thiazolidinediones
NETsNeutrophil extracellular traps
BSL3Biosafety level 3
RBDReceptor binding domain
VLPVirus-like particle
AdVAdenovirus
LNPLipid nanoparticle
RDVRemdesivir
RdRps RNA-dependent RNA polymerases
FDAFood and Drug Administration
IVIntravenous
HCVHepatitis C virus
LPVLopinavir
HIVHuman immunodeficiency virus
CYP3A4Cytochrome P450 3A4
3CLpro3C-like protease
ELACOIEfficacy of Lopinavir Plus Ritonavir and Arbidol Against Novel Coronavirus Infection
RECOVERYRandomized Evaluation of COVID-19 Therapy
IFNInterferon
HAHemagglutinin
HCQHydroxychloroquine
CQChloroquine
CPConvalescent plasma
NAbsNeutralizing Abs
CRSCytokine release syndrome
IL-6RIL-6 receptor
JAKJanus kinase
Tyk2Tyrosine kinase 2
STATSignal transducer and activator of transcription
RARheumatoid arthritis
AAK1Adaptor-associated protein kinase 1
AP2Adaptor protein-2 complex
HMG-CoAHydroxymethyl-glutaryl coenzyme A
OACsOral anticoagulants
GLP-1R GLP-1 receptor
SARS-CoV-2 MAMouse-adapted SARS-CoV-2
DIO Diet-induced-obese

References

  1. Yan, R.; Zhang, Y.; Li, Y.; Xia, L.; Guo, Y.; Zhou, Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science 2020, 367, 1444–1448. [Google Scholar] [CrossRef] [Green Version]
  2. Wölfel, R.; Corman, V.M.; Guggemos, W.; Seilmaier, M.; Zange, S.; Müller, M.A.; Niemeyer, D.; Jones, T.C.; Vollmar, P.; Rothe, C.; et al. Virological assessment of hospitalized patients with COVID-2019. Nat. Cell Biol. 2020, 581, 465–469. [Google Scholar] [CrossRef] [Green Version]
  3. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Franceschi, C.; Bonafè, M.; Valensin, S.; Olivieri, F.; de Luca, M.; Ottaviani, E.; de Benedictis, G. Inflamm-aging: An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef]
  5. Franceschi, C.; Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2014, 69 (Suppl. 1), S4–S9. [Google Scholar] [CrossRef]
  6. Campisi, J.; Daddadifagagna, F. Cellular senescence: When bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 2007, 8, 729–740. [Google Scholar] [CrossRef] [PubMed]
  7. Franceschi, C.; Bonafè, M.; Valensin, S. Human immunosenescence: The prevailing of innate immunity, the failing of clonotypic immunity, and the filling of immunological space. Vaccine 2000, 18, 1717–1720. [Google Scholar] [CrossRef]
  8. Shaw, A.C.; Joshi, S.; Greenwood, H.; Panda, A.; Lord, J.M. Aging of the innate immune system. Curr. Opin. Immunol. 2010, 22, 507–513. [Google Scholar] [CrossRef] [Green Version]
  9. Biagi, E.; Candela, M.; Franceschi, C.; Brigidi, P. The aging gut microbiota: New perspectives. Ageing Res. Rev. 2011, 10, 428–429. [Google Scholar] [CrossRef]
  10. Frasca, D.; Blomberg, B.B.; Paganelli, R. Aging, Obesity, and Inflammatory Age-Related Diseases. Front. Immunol. 2017, 8, 1745. [Google Scholar] [CrossRef] [PubMed]
  11. Grundy, S.M.; Brewer, H.B., Jr.; Cleeman, J.I.; Smith, S.C., Jr.; Lenfant, C.; American Heart, A.; National Heart, L.; Blood, I. Definition of Metabolic Syndrome: Report of the National Heart, Lung, and Blood Institute/American Heart Association conference on scientific issues related to definition. Circulation 2004, 109, 433–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Saltiel, A.R.; Olefsky, J.M. Inflammatory mechanisms linking obesity and metabolic disease. J. Clin. Investig. 2017, 127, 1–4. [Google Scholar] [CrossRef]
  13. Di Angelantonio, E.; Bhupathiraju, S.N.; Wormser, D.; Gao, P.; Kaptoge, S.; de Gonzalez, A.B.; Cairns, B.; Huxley, R.; Jackson, C.; Joshy, G.; et al. Body-mass index and all-cause mortality: Individual-participant-data meta-analysis of 239 prospective studies in four continents. Lancet 2016, 388, 776–786. [Google Scholar] [CrossRef] [Green Version]
  14. Fontaine, K.R.; Redden, D.T.; Wang, C.; Westfall, A.O.; Allison, D. Years of Life Lost Due to Obesity. JAMA 2003, 289, 187–193. [Google Scholar] [CrossRef] [PubMed]
  15. Tam, B.T.; Morais, J.A.; Santosa, S. Obesity and ageing: Two sides of the same coin. Obes. Rev. 2020, 21, e12991. [Google Scholar] [CrossRef] [PubMed]
  16. Leng, J.; Goldstein, D.R. Impact of aging on viral infections. Microbes Infect. 2010, 12, 1120–1124. [Google Scholar] [CrossRef] [Green Version]
  17. Andersen, C.J.; E Murphy, K.; Fernandez, M.L. Impact of Obesity and Metabolic Syndrome on Immunity. Adv. Nutr. 2016, 7, 66–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Honce, R.; Schultz-Cherry, S. Impact of Obesity on Influenza A Virus Pathogenesis, Immune Response, and Evolution. Front. Immunol. 2019, 10, 1071. [Google Scholar] [CrossRef] [PubMed]
  19. Smith, M.; Honce, R.; Schultz-Cherry, S. Metabolic Syndrome and Viral Pathogenesis: Lessons from Influenza and Coronaviruses. J. Virol. 2020, 94. [Google Scholar] [CrossRef]
  20. Santesmasses, D.; Castro, J.P.; Zenin, A.; Shindyapina, A.; Gerashchenko, M.; Zhang, B.; Kerepesi, C.; Yim, S.H.; Fedichev, P.O.; Gladyshev, V.N. COVID-19 is an emergent disease of aging. Aging Cell 2020, 19. [Google Scholar] [CrossRef] [PubMed]
  21. Geller, C.; Varbanov, M.; Duval, R.E. Human Coronaviruses: Insights into Environmental Resistance and Its Influence on the Development of New Antiseptic Strategies. Viruses 2012, 4, 3044–3068. [Google Scholar] [CrossRef] [Green Version]
  22. Ward, H.; Atchison, C.; Whitaker, M.; Ainslie, K.E.C.; Elliott, J.; Okell, L.; Redd, R.; Ashby, D.; Donnelly, C.A.; Barclay, W.; et al. SARS-CoV-2 antibody prevalence in England following the first peak of the pandemic. Nat. Commun. 2021, 12, 1–8. [Google Scholar] [CrossRef]
  23. Pastor-Barriuso, R.; Pérez-Gómez, B.; Hernán, M.A.; Pérez-Olmeda, M.; Yotti, R.; Oteo-Iglesias, J.; Sanmartín, J.L.; León-Gómez, I.; Fernández-García, A.; Fernández-Navarro, P.; et al. Infection fatality risk for SARS-CoV-2 in community dwelling population of Spain: Nationwide seroepidemiological study. BMJ 2020, 371, m4509. [Google Scholar] [CrossRef]
  24. O’Driscoll, M.; Ribeiro Dos Santos, G.; Wang, L.; Cummings, D.A.T.; Azman, A.S.; Paireau, J.; Fontanet, A.; Cauchemez, S.; Salje, H. Age-specific mortality and immunity patterns of SARS-CoV-2. Nat. Cell Biol. 2021, 590, 140–145. [Google Scholar] [CrossRef]
  25. Mueller, A.L.; McNamara, M.; Sinclair, D.A. Why does COVID-19 disproportionately affect older people? Aging 2020, 12, 9959–9981. [Google Scholar] [CrossRef] [PubMed]
  26. Kovacs, E.J.; Boe, D.M.; Boule, L.A.; Curtis, B.J. Inflammaging and the Lung. Clin. Geriatr. Med. 2017, 33, 459–471. [Google Scholar] [CrossRef]
  27. Liao, M.; Liu, Y.; Yuan, J.; Wen, Y.; Xu, G.; Zhao, J.; Cheng, L.; Li, J.; Wang, X.; Wang, F.; et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 2020, 26, 842–844. [Google Scholar] [CrossRef] [PubMed]
  28. Diao, B.; Wang, C.; Tan, Y.; Chen, X.; Liu, Y.; Ning, L.; Chen, L.; Li, M.; Liu, Y.; Wang, G.; et al. Reduction and Functional Exhaustion of T Cells in Patients with Coronavirus Disease 2019 (COVID-19). Front. Immunol. 2020, 11, 827. [Google Scholar] [CrossRef]
  29. Weaver, L.K.; Behrens, E.M. Weathering the Storm: Improving Therapeutic Interventions for Cytokine Storm Syndromes by Targeting Disease Pathogenesis. Curr. Treat. Options Rheumatol. 2017, 3, 33–48. [Google Scholar] [CrossRef] [Green Version]
  30. Sanada, F.; Taniyama, Y.; Muratsu, J.; Otsu, R.; Shimizu, H.; Rakugi, H.; Morishita, R. Source of Chronic Inflammation in Aging. Front. Cardiovasc. Med. 2018, 5, 12. [Google Scholar] [CrossRef] [Green Version]
  31. Buford, T.W.; Carter, C.S.; Vanderpol, W.J.; Chen, D.; Lefkowitz, E.J.; Eipers, P.; Morrow, C.D.; Bamman, M.M. Composition and richness of the serum microbiome differ by age and link to systemic inflammation. GeroScience 2018, 40, 257–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Mirsoian, A.; Bouchlaka, M.N.; Sckisel, G.D.; Chen, M.; Pai, C.-C.S.; Maverakis, E.; Spencer, R.G.; Fishbein, K.W.; Siddiqui, S.; Monjazeb, A.M.; et al. Adiposity induces lethal cytokine storm after systemic administration of stimulatory immunotherapy regimens in aged mice. J. Exp. Med. 2014, 211, 2373–2383. [Google Scholar] [CrossRef]
  33. Krabbe, K.S.; Pedersen, M.; Bruunsgaard, H. Inflammatory mediators in the elderly. Exp. Gerontol. 2004, 39, 687–699. [Google Scholar] [CrossRef] [PubMed]
  34. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
  35. Stout-Delgado, H.W.; Cho, S.J.; Chu, S.G.; Mitzel, D.N.; Villalba, J.; El-Chemaly, S.; Ryter, S.W.; Choi, A.M.K.; Rosas, I.O. Age-Dependent Susceptibility to Pulmonary Fibrosis Is Associated with NLRP3 Inflammasome Activation. Am. J. Respir. Cell Mol. Biol. 2016, 55, 252–263. [Google Scholar] [CrossRef] [Green Version]
  36. Zhao, C.; Zhao, W. NLRP3 Inflammasome—A Key Player in Antiviral Responses. Front. Immunol. 2020, 11, 211. [Google Scholar] [CrossRef] [Green Version]
  37. He, M.; Chiang, H.-H.; Luo, H.; Zheng, Z.; Qiao, Q.; Wang, L.; Tan, M.; Ohkubo, R.; Mu, W.-C.; Zhao, S.; et al. An Acetylation Switch of the NLRP3 Inflammasome Regulates Aging-Associated Chronic Inflammation and Insulin Resistance. Cell Metab. 2020, 31, 580–591.e5. [Google Scholar] [CrossRef] [PubMed]
  38. Massudi, H.; Grant, R.; Braidy, N.; Guest, J.; Farnsworth, B.; Guillemin, G.J. Age-Associated Changes in Oxidative Stress and NAD+ Metabolism In Human Tissue. PLoS ONE 2012, 7, e42357. [Google Scholar] [CrossRef] [PubMed]
  39. Heer, C.D.; Sanderson, D.J.; Voth, L.S.; Alhammad, Y.M.; Schmidt, M.S.; Trammell, S.A.; Perlman, S.; Cohen, M.S.; Fehr, A.R.; Brenner, C. Coronavirus infection and PARP expression dysregulate the NAD metabolome: An actionable component of innate immunity. J. Biol. Chem. 2020, 295, 17986–17996. [Google Scholar] [CrossRef]
  40. Ahmed, N. Advanced glycation end products—role in pathology of diabetic complications. Diabetes Res. Clin. Pract. 2005, 67, 3–21. [Google Scholar] [CrossRef]
  41. Singh, R.; Barden, A.; Mori, T.; Beilin, L. Advanced glycation end-products: A review. Diabetol. 2001, 44, 129–146. [Google Scholar] [CrossRef] [Green Version]
  42. Chen, I.-Y.; Moriyama, M.; Chang, M.-F.; Ichinohe, T. Severe Acute Respiratory Syndrome Coronavirus Viroporin 3a Activates the NLRP3 Inflammasome. Front. Microbiol. 2019, 10, 50. [Google Scholar] [CrossRef] [Green Version]
  43. Son, S.; Hwang, I.; Han, S.H.; Shin, J.-S.; Shin, O.S.; Yu, J.-W. Advanced glycation end products impair NLRP3 inflammasome–mediated innate immune responses in macrophages. J. Biol. Chem. 2017, 292, 20437–20448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Mannick, J.B.; Del Giudice, G.; Lattanzi, M.; Valiante, N.M.; Praestgaard, J.; Huang, B.; Lonetto, M.A.; Maecker, H.T.; Kovarik, J.; Carson, S.; et al. mTOR inhibition improves immune function in the elderly. Sci. Transl. Med. 2014, 6, 268ra179. [Google Scholar] [CrossRef]
  45. Mannick, J.B.; Morris, M.; Hockey, H.-U.P.; Roma, G.; Beibel, M.; Kulmatycki, K.; Watkins, M.; Shavlakadze, T.; Zhou, W.; Quinn, D.; et al. TORC1 inhibition enhances immune function and reduces infections in the elderly. Sci. Transl. Med. 2018, 10, eaaq1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Sharma, S.; Ray, A.; Sadasivam, B. Metformin in COVID-19: A possible role beyond diabetes. Diabetes Res. Clin. Pr. 2020, 164, 108183. [Google Scholar] [CrossRef] [PubMed]
  47. Bansal, R.; Gubbi, S.; Muniyappa, R. Metabolic Syndrome and COVID 19: Endocrine-Immune-Vascular Interactions Shapes Clinical Course. Endocrinol. 2020, 161. [Google Scholar] [CrossRef]
  48. Cai, Q.; Chen, F.; Wang, T.; Luo, F.; Liu, X.; Wu, Q.; He, Q.; Wang, Z.; Liu, Y.; Liu, L.; et al. Obesity and COVID-19 Severity in a Designated Hospital in Shenzhen, China. Diabetes Care 2020, 43, 1392–1398. [Google Scholar] [CrossRef] [PubMed]
  49. Simonnet, A.; Chetboun, M.; Poissy, J.; Raverdy, V.; Noulette, J.; Duhamel, A.; Labreuche, J.; Mathieu, D.; Pattou, F.; Jourdain, M.; et al. High Prevalence of Obesity in Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) Requiring Invasive Mechanical Ventilation. Obesity 2020, 28, 1195–1199. [Google Scholar] [CrossRef]
  50. Williamson, E.J.; Walker, A.J.; Bhaskaran, K.; Bacon, S.; Bates, C.; Morton, C.E.; Curtis, H.J.; Mehrkar, A.; Evans, D.; Inglesby, P.; et al. Factors associated with COVID-19-related death using OpenSAFELY. Nature 2020, 584, 430–436. [Google Scholar] [CrossRef]
  51. Ritter, A.; Kreis, N.-N.; Louwen, F.; Yuan, J. Obesity and COVID-19: Molecular Mechanisms Linking both Pandemics. Int. J. Mol. Sci. 2020, 21, 5793. [Google Scholar] [CrossRef] [PubMed]
  52. Exley, M.A.; Hand, L.; O’Shea, D.; Lynch, L. Interplay between the immune system and adipose tissue in obesity. J. Endocrinol. 2014, 223, R41–R48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Cildir, G.; Akıncılar, S.C.; Tergaonkar, V. Chronic adipose tissue inflammation: All immune cells on the stage. Trends Mol. Med. 2013, 19, 487–500. [Google Scholar] [CrossRef]
  54. Huh, J.Y.; Park, Y.J.; Ham, M.; Kim, J.B. Crosstalk between Adipocytes and Immune Cells in Adipose Tissue Inflammation and Metabolic Dysregulation in Obesity. Mol. Cells 2014, 37, 365–371. [Google Scholar] [CrossRef]
  55. Louwen, F.; Ritter, A.; Kreis, N.N.; Yuan, J. Insight into the development of obesity: Functional alterations of adipose-derived mesenchymal stem cells. Obes. Rev. 2018, 19, 888–904. [Google Scholar] [CrossRef]
  56. Ritter, A.; Louwen, F.; Yuan, J. Deficient primary cilia in obese adipose-derived mesenchymal stem cells: Obesity, a secondary ciliopathy? Obes. Rev. 2018, 19, 1317–1328. [Google Scholar] [CrossRef]
  57. Parameswaran, K.; Todd, D.C.; Soth, M. Altered Respiratory Physiology in Obesity. Can. Respir. J. 2006, 13, 203–210. [Google Scholar] [CrossRef]
  58. Wang, J.; Wang, B.J.; Yang, J.C.; Wang, M.Y.; Chen, C.; Luo, G.X.; He, W.F. Research advances in the mechanism of pulmonary fibrosis induced by coronavirus disease 2019 and the corresponding therapeutic measures. Zhonghua Shao Shang Za Zhi 2020, 36, 691–697. [Google Scholar] [CrossRef] [PubMed]
  59. Fu, J.; Zhou, B.; Zhang, L.; Balaji, K.S.; Wei, C.; Liu, X.; Chen, H.; Peng, J.; Fu, J. Expressions and significances of the angiotensin-converting enzyme 2 gene, the receptor of SARS-CoV-2 for COVID-19. Mol. Biol. Rep. 2020, 47, 4383–4392. [Google Scholar] [CrossRef] [PubMed]
  60. Kruglikov, I.L.; Scherer, P.E. The Role of Adipocytes and Adipocyte-Like Cells in the Severity of COVID-19 Infections. Obesity 2020, 28, 1187–1190. [Google Scholar] [CrossRef]
  61. Lamers, D.; Famulla, S.; Wronkowitz, N.; Hartwig, S.; Lehr, S.; Ouwens, D.M.; Eckardt, K.; Kaufman, J.M.; Ryden, M.; Müller, S.; et al. Dipeptidyl Peptidase 4 Is a Novel Adipokine Potentially Linking Obesity to the Metabolic Syndrome. Diabetes 2011, 60, 1917–1925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Yanagimachi, T.; Fujita, Y.; Takeda, Y.; Honjo, J.; Sakagami, H.; Kitsunai, H.; Takiyama, Y.; Abiko, A.; Makino, Y.; Kieffer, T.J.; et al. Dipeptidyl peptidase-4 inhibitor treatment induces a greater increase in plasma levels of bioactive GIP than GLP-1 in non-diabetic subjects. Mol. Metab. 2017, 6, 226–231. [Google Scholar] [CrossRef]
  63. Sell, H.; Blüher, M.; Klöting, N.; Schlich, R.; Willems, M.; Ruppe, F.; Knoefel, W.T.; Dietrich, A.; Fielding, B.A.; Arner, P.; et al. Adipose Dipeptidyl Peptidase-4 and Obesity: Correlation with insulin resistance and depot-specific release from adipose tissue in vivo and in vitro. Diabetes Care 2013, 36, 4083–4090. [Google Scholar] [CrossRef] [Green Version]
  64. Shin, K.; Pandey, A.; Liu, X.-Q.; Anini, Y.; Rainey, J.K. Preferential apelin-13 production by the proprotein convertase PCSK3 is implicated in obesity. FEBS Open Bio. 2013, 3, 328–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Radzikowska, U.; Ding, M.; Tan, G.; Zhakparov, D.; Peng, Y.; Wawrzyniak, P.; Wang, M.; Li, S.; Morita, H.; Altunbulakli, C.; et al. Distribution of ACE2, CD147, CD26, and other SARS-CoV-2 associated molecules in tissues and immune cells in health and in asthma, COPD, obesity, hypertension, and COVID-19 risk factors. Allergy 2020, 75. [Google Scholar] [CrossRef] [PubMed]
  66. Cyranoski, D. Profile of a killer: The complex biology powering the coronavirus pandemic. Nat. Cell Biol. 2020, 581, 22–26. [Google Scholar] [CrossRef]
  67. Wang, K.; Chen, W.; Zhang, Z.; Deng, Y.; Lian, J.-Q.; Du, P.; Wei, D.; Zhang, Y.; Sun, X.-X.; Gong, L.; et al. CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal Transduct. Target. Ther. 2020, 5, 1–10. [Google Scholar] [CrossRef] [PubMed]
  68. Atawia, R.T.; Bunch, K.L.; Toque, H.A.; Caldwell, R.B.; Caldwell, R.W. Mechanisms of obesity-induced metabolic and vascular dysfunctions. Front. Biosci. 2019, 24, 890–934. [Google Scholar]
  69. Zhou, L.; Niu, Z.; Jiang, X.; Zhang, Z.; Zheng, Y.; Wang, Z.; Zhu, Y.; Gao, L.; Huang, H.; Wang, X.; et al. SARS-CoV-2 Targets by the pscRNA Profiling of ACE2, TMPRSS2 and Furin Proteases. iScience 2020, 23, 101744. [Google Scholar] [CrossRef] [PubMed]
  70. Tsao, P.S.; McEvoy, L.M.; Drexler, H.; Butcher, E.C.; Cooke, J. Enhanced endothelial adhesiveness in hypercholesterolemia is attenuated by L-arginine. Circulation 1994, 89, 2176–2182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Tsao, P.S.; Wang, B.-Y.; Buitrago, R.; Shyy, J.Y.-J.; Cooke, J.P. Nitric Oxide Regulates Monocyte Chemotactic Protein-1. Circulation 1997, 96, 934–940. [Google Scholar] [CrossRef]
  72. Jang, J.E.; Ko, M.S.; Yun, J.-Y.; Kim, M.-O.; Kim, J.H.; Park, H.S.; Kim, A.-R.; Kim, H.-J.; Kim, B.J.; Ahn, Y.E.; et al. Nitric Oxide Produced by Macrophages Inhibits Adipocyte Differentiation and Promotes Profibrogenic Responses in Preadipocytes to Induce Adipose Tissue Fibrosis. Diabetes 2016, 65, 2516–2528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Savini, I.; Catani, M.V.; Evangelista, D.; Gasperi, V.; Avigliano, L. Obesity-Associated Oxidative Stress: Strategies Finalized to Improve Redox State. Int. J. Mol. Sci. 2013, 14, 10497–10538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Könner, A.C.; Brüning, J.C. Toll-like receptors: Linking inflammation to metabolism. Trends Endocrinol. Metab. 2011, 22, 16–23. [Google Scholar] [CrossRef] [PubMed]
  75. Artemniak-Wojtowicz, D.; Kucharska, A.; Pyrżak, B. Obesity and chronic inflammation crosslinking. Central Eur. J. Immunol. 2020, 45, 461–468. [Google Scholar] [CrossRef]
  76. Ives, A.; Nomura, J.; Martinon, F.; Roger, T.; Le Roy, D.; Miner, J.N.; Simon, G.; Busso, N.; So, A. Xanthine oxidoreductase regulates macrophage IL1β secretion upon NLRP3 inflammasome activation. Nat. Commun. 2015, 6, 6555. [Google Scholar] [CrossRef] [Green Version]
  77. Gainsford, T.; Willson, T.A.; Metcalf, D.; Handman, E.; McFarlane, C.; Ng, A.; Nicola, N.A.; Alexander, W.S.; Hilton, D. Leptin can induce proliferation, differentiation, and functional activation of hemopoietic cells. Proc. Natl. Acad. Sci. USA 1996, 93, 14564–14568. [Google Scholar] [CrossRef] [Green Version]
  78. Green, W.D.; Beck, M.A. Obesity Impairs the Adaptive Immune Response to Influenza Virus. Ann. Am. Thorac. Soc. 2017, 14, S406–S409. [Google Scholar] [CrossRef] [PubMed]
  79. Neidich, S.D.; Green, W.D.; Rebeles, J.; Karlsson, E.A.; Schultz-Cherry, S.; Noah, T.L.; Chakladar, S.; Hudgens, M.G.; Weir, S.S.; A Beck, M. Increased risk of influenza among vaccinated adults who are obese. Int. J. Obes. 2017, 41, 1324–1330. [Google Scholar] [CrossRef] [Green Version]
  80. Sheridan, P.A.; Paich, H.A.; Handy, J.A.; Karlsson, E.; Hudgens, M.G.; Sammon, A.B.; Holland, L.A.; Weir, S.; Noah, T.L.; Beck, M.A. Obesity is associated with impaired immune response to influenza vaccination in humans. Int. J. Obes. 2011, 36, 1072–1077. [Google Scholar] [CrossRef] [Green Version]
  81. Paich, H.A.; Sheridan, P.A.; Handy, J.; Karlsson, E.A.; Schultz-Cherry, S.; Hudgens, M.G.; Noah, T.L.; Weir, S.S.; Beck, M.A. Overweight and obese adult humans have a defective cellular immune response to pandemic H1N1 Influenza a virus. Obesity 2013, 21, 2377–2386. [Google Scholar] [CrossRef] [PubMed]
  82. Poirier, P.; Giles, T.D.; Bray, G.A.; Hong, Y.; Stern, J.S.; Pi-Sunyer, F.X.; Eckel, R.H. Obesity and Cardiovascular Disease: Pathophysiology, Evaluation, and Effect of Weight Loss: An update of the 1997 American Heart Association Scientific Statement on Obesity and Heart Disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation 2006, 113, 898–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Madjid, M.; Miller, C.C.; Zarubaev, V.V.; Marinich, I.G.; Kiselev, O.I.; Lobzin, Y.; Filippov, A.E.; Casscells, S.W. Influenza epidemics and acute respiratory disease activity are associated with a surge in autopsy-confirmed coronary heart disease death: Results from 8 years of autopsies in 34,892 subjects. Eur. Hear. J. 2007, 28, 1205–1210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kwong, J.C.; Schwartz, K.L.; Campitelli, M.A.; Chung, H.; Crowcroft, N.S.; Karnauchow, T.; Katz, K.; Ko, D.; McGeer, A.J.; McNally, D.; et al. Acute Myocardial Infarction after Laboratory-Confirmed Influenza Infection. N. Engl. J. Med. 2018, 378, 345–353. [Google Scholar] [CrossRef] [PubMed]
  85. Madjid, M.; Connolly, A.T.; Nabutovsky, Y.; Safavi-Naeini, P.; Razavi, M.; Miller, C.C. Effect of High Influenza Activity on Risk of Ventricular Arrhythmias Requiring Therapy in Patients with Implantable Cardiac Defibrillators and Cardiac Resynchronization Therapy Defibrillators. Am. J. Cardiol. 2019, 124, 44–50. [Google Scholar] [CrossRef]
  86. Kytömaa, S.; Hegde, S.; Claggett, B.; Udell, J.A.; Rosamond, W.; Temte, J.; Nichol, K.; Wright, J.D.; Solomon, S.D.; Vardeny, O. Association of Influenza-like Illness Activity with Hospitalizations for Heart Failure. JAMA Cardiol. 2019, 4, 363–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Vardeny, O.; Solomon, S.D. Influenza vaccination: A one-shot deal to reduce cardiovascular events. Eur. Hear. J. 2016, 38, 334–337. [Google Scholar] [CrossRef]
  88. Madjid, M.; Aboshady, I.; Awan, I.; Litovsky, S.; Casscells, S.W. Influenza and cardiovascular disease: Is there a causal relationship? Tex. Heart Inst. J. 2004, 31, 4–13. [Google Scholar]
  89. Corrales-Medina, V.F.; Madjid, M.; Musher, D.M. Role of acute infection in triggering acute coronary syndromes. Lancet Infect. Dis. 2010, 10, 83–92. [Google Scholar] [CrossRef]
  90. Madjid, M.; Safavi-Naeini, P.; Solomon, S.D.; Vardeny, O. Potential Effects of Coronaviruses on the Cardiovascular System. JAMA Cardiol. 2020, 5, 831. [Google Scholar] [CrossRef] [Green Version]
  91. Bansal, M. Cardiovascular disease and COVID-19. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 247–250. [Google Scholar] [CrossRef] [PubMed]
  92. Xiong, T.-Y.; Redwood, S.; Prendergast, B.; Chen, M. Coronaviruses and the cardiovascular system: Acute and long-term implications. Eur. Hear. J. 2020, 41, 1798–1800. [Google Scholar] [CrossRef] [Green Version]
  93. Li, B.; Yang, J.; Zhao, F.; Zhi, L.; Wang, X.; Liu, L.; Bi, Z.; Zhao, Y. Prevalence and impact of cardiovascular metabolic diseases on COVID-19 in China. Clin. Res. Cardiol. 2020, 109, 531–538. [Google Scholar] [CrossRef]
  94. 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, 5, 811–818. [Google Scholar] [CrossRef] [Green Version]
  95. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  96. Schiavone, M.; Gobbi, C.; Biondi-Zoccai, G.; D’Ascenzo, F.; Palazzuoli, A.; Gasperetti, A.; Mitacchione, G.; Viecca, M.; Galli, M.; Fedele, F.; et al. Acute Coronary Syndromes and Covid-19: Exploring the Uncertainties. J. Clin. Med. 2020, 9, 1683. [Google Scholar] [CrossRef]
  97. Shi, S.; Qin, M.; Shen, B.; Cai, Y.; Liu, T.; Yang, F.; Gong, W.; Liu, X.; Liang, J.; Zhao, Q.; et al. Association of Cardiac Injury with Mortality in Hospitalized Patients With COVID-19 in Wuhan, China. JAMA Cardiol. 2020, 5, 802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Chen, C.; Cohrs, C.M.; Stertmann, J.; Bozsak, R.; Speier, S. Human beta cell mass and function in diabetes: Recent advances in knowledge and technologies to understand disease pathogenesis. Mol. Metab. 2017, 6, 943–957. [Google Scholar] [CrossRef]
  99. Christensen, A.; Gannon, M. The Beta Cell in Type 2 Diabetes. Curr. Diabetes Rep. 2019, 19, 81. [Google Scholar] [CrossRef]
  100. Tabák, A.G.; Jokela, M.; Akbaraly, T.N.; Brunner, E.J.; Kivimäki, M.; Witte, D.R. Trajectories of glycaemia, insulin sensitivity, and insulin secretion before diagnosis of type 2 diabetes: An analysis from the Whitehall II study. Lancet 2009, 373, 2215–2221. [Google Scholar] [CrossRef] [Green Version]
  101. Zhu, L.; She, Z.-G.; Cheng, X.; Qin, J.-J.; Zhang, X.-J.; Cai, J.; Lei, F.; Wang, H.; Xie, J.; Wang, W.; et al. Association of Blood Glucose Control and Outcomes in Patients with COVID-19 and Pre-existing Type 2 Diabetes. Cell Metab. 2020, 31, 1068–1077.e3. [Google Scholar] [CrossRef] [PubMed]
  102. Cariou, B.; Hadjadj, S.; Wargny, M.; Pichelin, M.; Al-Salameh, A.; Allix, I.; Amadou, C.; Arnault, G.; Baudoux, F.; Bauduceau, B.; et al. Phenotypic characteristics and prognosis of inpatients with COVID-19 and diabetes: The CORONADO study. Diabetologia 2020, 63, 1500–1515. [Google Scholar] [CrossRef]
  103. Muniyappa, R.; Gubbi, S. COVID-19 pandemic, coronaviruses, and diabetes mellitus. Am. J. Physiol. Metab. 2020, 318, E736–E741. [Google Scholar] [CrossRef] [Green Version]
  104. Fernandez, C.; Rysä, J.; Almgren, P.; Nilsson, J.; Engström, G.; Orho-Melander, M.; Ruskoaho, H.; Melander, O. Plasma levels of the proprotein convertase furin and incidence of diabetes and mortality. J. Intern. Med. 2018, 284, 377–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Roca-Ho, H.; Riera, M.; Palau, V.; Pascual, J.; Soler, M.J. Characterization of ACE and ACE2 Expression within Different Organs of the NOD Mouse. Int. J. Mol. Sci. 2017, 18, 563. [Google Scholar] [CrossRef] [Green Version]
  106. Wysocki, J.; Ye, M.; Soler, M.J.; Gurley, S.B.; Xiao, H.D.; Bernstein, K.; Coffman, T.M.; Chen, S.; Batlle, D. ACE and ACE2 Activity in Diabetic Mice. Diabetes 2006, 55, 2132–2139. [Google Scholar] [CrossRef] [Green Version]
  107. Zou, X.; Chen, K.; Zou, J.; Han, P.; Hao, J.; Han, Z. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to 2019-nCoV infection. Front. Med. 2020, 14, 185–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Asperen, R.M.W.-V.; Bos, A.P.; Bem, R.A.; Dierdorp, B.S.; Dekker, T.; van Goor, H.; Kamilic, J.; van der Loos, C.M.; Berg, E.V.D.; Bruijn, M.; et al. Imbalance Between Pulmonary Angiotensin-Converting Enzyme and Angiotensin-Converting Enzyme 2 Activity in Acute Respiratory Distress Syndrome. Pediatr. Crit. Care Med. 2013, 14, e438–e441. [Google Scholar] [CrossRef] [PubMed]
  109. Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B.; Huan, Y.; Yang, P.; Zhang, Y.; Deng, W.; et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus–induced lung injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef]
  110. Monteil, V.; Kwon, H.; Prado, P.; Hagelkrüys, A.; Wimmer, R.A.; Stahl, M.; Leopoldi, A.; Garreta, E.; Del Pozo, C.H.; Prosper, F.; et al. Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell 2020, 181, 905–913.e7. [Google Scholar] [CrossRef]
  111. Tsalamandris, S.; Antonopoulos, A.; Oikonomou, E.; Papamikroulis, G.-A.; Vogiatzi, G.; Papaioannou, S.; Deftereos, S.; Tousoulis, D. The Role of Inflammation in Diabetes: Current Concepts and Future Perspectives. Eur. Cardiol. Rev. 2019, 14, 50–59. [Google Scholar] [CrossRef] [Green Version]
  112. Erener, S. Diabetes, infection risk and COVID-19. Mol. Metab. 2020, 39, 101044. [Google Scholar] [CrossRef]
  113. Sena, C.M.; Pereira, A.M.; Seiça, R. Endothelial dysfunction—A major mediator of diabetic vascular disease. Biochim. Biophys. Acta Mol. Basis Dis. 2013, 1832, 2216–2231. [Google Scholar] [CrossRef] [Green Version]
  114. Madhusudhan, T.; Ruf, W. Coagulation Signalling and Metabolic Disorders: Lessons Learned from Animal Models. Hämostaseologie 2019, 39, 164–172. [Google Scholar] [CrossRef]
  115. Kearney, K.; Tomlinson, D.; Smith, K.; Ajjan, R. Hypofibrinolysis in diabetes: A therapeutic target for the reduction of cardiovascular risk. Cardiovasc. Diabetol. 2017, 16, 1–17. [Google Scholar] [CrossRef] [Green Version]
  116. Jenny, L.; Melmer, A.; Laimer, M.; Hardy, E.T.; Lam, W.A.; Schroeder, V. Diabetes affects endothelial cell function and alters fibrin clot formation in a microvascular flow model: A pilot study. Diabetes Vasc. Dis. Res. 2020, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Berezin, A. Neutrophil extracellular traps: The core player in vascular complications of diabetes mellitus. Diabetes Metab. Syndr. Clin. Res. Rev. 2019, 13, 3017–3023. [Google Scholar] [CrossRef] [PubMed]
  118. Carestia, A.; Frechtel, G.; Cerrone, G.; Linari, M.A.; Gonzalez, C.D.; Casais, P.; Schattner, M. NETosis before and after Hyperglycemic Control in Type 2 Diabetes Mellitus Patients. PLoS ONE 2016, 11, e0168647. [Google Scholar] [CrossRef] [PubMed]
  119. Connors, J.M.; Levy, J.H. COVID-19 and its implications for thrombosis and anticoagulation. Blood 2020, 135, 2033–2040. [Google Scholar] [CrossRef] [PubMed]
  120. Sardu, C.; D’Onofrio, N.; Balestrieri, M.L.; Barbieri, M.; Rizzo, M.R.; Messina, V.; Maggi, P.; Coppola, N.; Paolisso, G.; Marfella, R. Outcomes in Patients with Hyperglycemia Affected by COVID-19: Can We Do More on Glycemic Control? Diabetes Care 2020, 43, 1408–1415. [Google Scholar] [CrossRef] [PubMed]
  121. Krammer, F. SARS-CoV-2 vaccines in development. Nat. Cell Biol. 2020, 586, 1–16. [Google Scholar] [CrossRef] [PubMed]
  122. Ashraf, M.; Kim, Y.; Kumar, S.; Seo, D.; Ashraf, M.; Bae, Y.-S. COVID-19 Vaccines (Revisited) and Oral-Mucosal Vector System as a Potential Vaccine Platform. Vaccines 2021, 9, 171. [Google Scholar] [CrossRef] [PubMed]
  123. Vellozzi, C.; Burwen, D.R.; Dobardzic, A.; Ball, R.; Walton, K.; Haber, P. Safety of trivalent inactivated influenza vaccines in adults: Background for pandemic influenza vaccine safety monitoring. Vaccine 2009, 27, 2114–2120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Murdin, A.D.; Barreto, L.; Plotkin, S. Inactivated poliovirus vaccine: Past and present experience. Vaccine 1996, 14, 735–746. [Google Scholar] [CrossRef]
  125. Kusov, Y.; Elbert, L.; Nelga, I.; Grishina, G.; Dunaevski, O.; Kharin, N.; Maslov, Y.; Drozdov, S.; Balayan, M. Immunogenicity trial of inactivated hepatitis A virus vaccine in human volunteers. Vaccine 1991, 9, 540–541. [Google Scholar] [CrossRef]
  126. Wu, W.; Liu, D.; Li, K.; Nuorti, J.P.; Nohynek, H.M.; Xu, D.; Ye, J.; Zheng, J.; Wang, H. Post-marketing safety surveillance for inactivated and live-attenuated Japanese encephalitis vaccines in China, 2008–2013. Vaccine 2017, 35, 3666–3671. [Google Scholar] [CrossRef] [Green Version]
  127. Qamar, M.T.U.; Saleem, S.; Ashfaq, U.A.; Bari, A.; Anwar, F.; Alqahtani, S. Epitope-based peptide vaccine design and target site depiction against Middle East Respiratory Syndrome Coronavirus: An immune-informatics study. J. Transl. Med. 2019, 17, 1–14. [Google Scholar] [CrossRef]
  128. Gao, Q.; Bao, L.; Mao, H.; Wang, L.; Xu, K.; Yang, M.; Li, Y.; Zhu, L.; Wang, N.; Lv, Z.; et al. Development of an inactivated vaccine candidate for SARS-CoV-2. Science 2020, 369, 77–81. [Google Scholar] [CrossRef]
  129. Wang, H.; Zhang, Y.; Huang, B.; Deng, W.; Quan, Y.; Wang, W.; Xu, W.; Zhao, Y.; Li, N.; Zhang, J.; et al. Development of an Inactivated Vaccine Candidate, BBIBP-CorV, with Potent Protection against SARS-CoV-2. Cell 2020, 182, 713–721.e9. [Google Scholar] [CrossRef]
  130. Minor, P.D. Live attenuated vaccines: Historical successes and current challenges. Virology 2015, 479, 379–392. [Google Scholar] [CrossRef] [Green Version]
  131. Mohn, K.G.-I.; Smith, I.; Sjursen, H.; Cox, R.J. Immune responses after live attenuated influenza vaccination. Hum. Vaccines Immunother. 2018, 14, 571–578. [Google Scholar] [CrossRef] [PubMed]
  132. Talon, J.; Salvatore, M.; O’Neill, R.E.; Nakaya, Y.; Zheng, H.; Muster, T.; García-Sastre, A.; Palese, P. Influenza A and B viruses expressing altered NS1 proteins: A vaccine approach. Proc. Natl. Acad. Sci. USA 2000, 97, 4309–4314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Broadbent, A.J.; Santos, C.P.; Anafu, A.; Wimmer, E.; Mueller, S.; Subbarao, K. Evaluation of the attenuation, immunogenicity, and efficacy of a live virus vaccine generated by codon-pair bias de-optimization of the 2009 pandemic H1N1 influenza virus, in ferrets. Vaccine 2016, 34, 563–570. [Google Scholar] [CrossRef] [Green Version]
  134. Griffin, D.E. Measles Vaccine. Viral Immunol. 2018, 31, 86–95. [Google Scholar] [CrossRef] [PubMed]
  135. Plotkin, S. History of vaccination. Proc. Natl. Acad. Sci. USA 2014, 111, 12283–12287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Guardeno, J.M.J.; Regla-Nava, J.A.; Nieto-Torres, J.L.; de Diego, M.L.; Castaño-Rodriguez, C.; Fernandez-Delgado, R.; Perlman, S.; Enjuanes, L. Identification of the Mechanisms Causing Reversion to Virulence in an Attenuated SARS-CoV for the Design of a Genetically Stable Vaccine. PLOS Pathog. 2015, 11, e1005215. [Google Scholar] [CrossRef] [Green Version]
  137. Live attenuated influenza vaccine for children. Drug Ther. Bull. 2017, 55, 114–117. [CrossRef]
  138. Armitage, E.P.; Camara, J.; Bah, S.; Forster, A.S.; Clarke, E.; Kampmann, B.; de Silva, T. Acceptability of intranasal live attenuated influenza vaccine, influenza knowledge and vaccine intent in The Gambia. Vaccine 2018, 36, 1772–1780. [Google Scholar] [CrossRef]
  139. Esposito, S.; Montinaro, V.; Groppali, E.; Tenconi, R.; Semino, M.; Principi, N. Live attenuated intranasal influenza vaccine. Hum. Vaccines Immunother. 2012, 8, 76–80. [Google Scholar] [CrossRef] [Green Version]
  140. Wang, J.; Peng, Y.; Xu, H.; Cui, Z.; Williams, R. The COVID-19 Vaccine Race: Challenges and Opportunities in Vaccine Formulation. AAPS Pharm. Sci. Tech. 2020, 21, 225. [Google Scholar] [CrossRef]
  141. 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, 12, 1–5. [Google Scholar] [CrossRef] [PubMed]
  142. Sakaguchi, W.; Kubota, N.; Shimizu, T.; Saruta, J.; Fuchida, S.; Kawata, A.; Yamamoto, Y.; Sugimoto, M.; Yakeishi, M.; Tsukinoki, K. Existence of SARS-CoV-2 Entry Molecules in the Oral Cavity. Int. J. Mol. Sci. 2020, 21, 6000. [Google Scholar] [CrossRef] [PubMed]
  143. Gurwith, M.; Condit, R.C.; Excler, J.-L.; Robertson, J.S.; Kim, D.; Fast, P.E.; Drew, S.; Wood, D.; Klug, B.; Whelan, M.; et al. Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) standardized template for collection of key information for benefit-risk assessment of live-attenuated viral vaccines. Vaccine 2020, 38, 7702–7707. [Google Scholar] [CrossRef]
  144. Seo, S.H.; Jang, Y. Cold-Adapted Live Attenuated SARS-Cov-2 Vaccine Completely Protects Human ACE2 Transgenic Mice from SARS-Cov-2 Infection. Vaccines 2020, 8, 584. [Google Scholar] [CrossRef]
  145. Amanat, F.; Stadlbauer, D.; Strohmeier, S.; Nguyen, T.H.O.; Chromikova, V.; McMahon, M.; Jiang, K.; Arunkumar, G.A.; Jurczyszak, D.; Polanco, J.; et al. A serological assay to detect SARS-CoV-2 seroconversion in humans. Nat. Med. 2020, 26, 1033–1036. [Google Scholar] [CrossRef]
  146. Chen, W.H.; Tao, X.; Agrawal, A.; Algaissi, A.; Peng, B.H.; Pollet, J.; Strych, U.; Bottazzi, M.E.; Hotez, P.J.; Lustigman, S.; et al. Yeast-Expressed SARS-CoV Recombinant Receptor-Binding Domain (RBD219-N1) Formulated with Alum Induces Protective Immunity and Reduces Immune Enhancement. bioRxiv 2020. [Google Scholar] [CrossRef]
  147. Chen, J.; Miao, L.; Li, J.-M.; Li, Y.-Y.; Zhu, Q.-Y.; Zhou, C.-L.; Fang, H.-Q.; Chen, H.-P. Receptor-binding domain of SARS-Cov spike protein: Soluble expression in E.coli, purification and functional characterization. World J. Gastroenterol. 2005, 11, 6159–6164. [Google Scholar] [CrossRef]
  148. Cox, M.M.; Hollister, J.R. FluBlok, a next generation influenza vaccine manufactured in insect cells. Biology 2009, 37, 182–189. [Google Scholar] [CrossRef] [PubMed]
  149. Cox, M.M.; Patriarca, P.A.; Treanor, J. FluBlok, a recombinant hemagglutinin influenza vaccine. Influ. Other Respir. Viruses 2008, 2, 211–219. [Google Scholar] [CrossRef] [Green Version]
  150. Cox, M.M.J.; Izikson, R.; Post, P.; Dunkle, L.M. Safety, efficacy, and immunogenicity of Flublok in the prevention of seasonal influenza in adults. Ther. Adv. Vaccines 2015, 3, 97–108. [Google Scholar] [CrossRef] [Green Version]
  151. Liu, Z.; Xu, W.; Xia, S.; Gu, C.; Wang, X.; Wang, Q.; Zhou, J.; Wu, Y.; Cai, X.; Qu, D.; et al. RBD-Fc-based COVID-19 vaccine candidate induces highly potent SARS-CoV-2 neutralizing antibody response. Signal Transduct. Target. Ther. 2020, 5, 1–10. [Google Scholar] [CrossRef] [PubMed]
  152. Keech, C.; Albert, G.; Cho, I.; Robertson, A.; Reed, P.; Neal, S.; Plested, J.S.; Zhu, M.; Cloney-Clark, S.; Zhou, H.; et al. Phase 1–2 Trial of a SARS-CoV-2 Recombinant Spike Protein Nanoparticle Vaccine. N. Engl. J. Med. 2020, 383, 2320–2332. [Google Scholar] [CrossRef]
  153. Zhu, F.-C.; Li, Y.-H.; Guan, X.-H.; Hou, L.-H.; Wang, W.-J.; Li, J.-X.; Wu, S.-P.; Wang, B.-S.; Wang, Z.; Wang, L.; et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: A dose-escalation, open-label, non-randomised, first-in-human trial. Lancet 2020, 395, 1845–1854. [Google Scholar] [CrossRef]
  154. Zhu, F.-C.; Guan, X.-H.; Li, Y.-H.; Huang, J.-Y.; Jiang, T.; Hou, L.-H.; Li, J.-X.; Yang, B.-F.; Wang, L.; Wang, W.-J.; et al. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2020, 396, 479–488. [Google Scholar] [CrossRef]
  155. Mercado, N.B.; Zahn, R.; Wegmann, F.; Loos, C.; Chandrashekar, A.; Yu, J.; Liu, J.; Peter, L.; Mcmahan, K.; Tostanoski, L.H.; et al. Single-shot Ad26 vaccine protects against SARS-CoV-2 in rhesus macaques. Nature 2020, 1–11. [Google Scholar] [CrossRef]
  156. Van Doremalen, N.; Lambe, T.; Spencer, A.; Belij-Rammerstorfer, S.; Purushotham, J.N.; Port, J.R.; Avanzato, V.A.; Bushmaker, T.; Flaxman, A.; Ulaszewska, M.; et al. ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia in rhesus macaques. Nature 2020, 586, 578–582. [Google Scholar] [CrossRef]
  157. Folegatti, P.M.; Ewer, K.J.; Aley, P.K.; Angus, B.; Becker, S.; Belij-Rammerstorfer, S.; Bellamy, D.; Bibi, S.; Bittaye, M.; Clutterbuck, E.A.; et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: A preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet 2020, 396, 467–478. [Google Scholar] [CrossRef]
  158. Logunov, D.Y.; Dolzhikova, I.V.; Zubkova, O.V.; Tukhvatullin, A.I.; Shcheblyakov, D.V.; Dzharullaeva, A.S.; Grousova, D.M.; Erokhova, A.S.; Kovyrshina, A.V.; Botikov, A.G.; et al. Safety and immunogenicity of an rAd26 and rAd5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: Two open, non-randomised phase 1/2 studies from Russia. Lancet 2020, 396, 887–897. [Google Scholar] [CrossRef]
  159. Argnani, R.; Lufino, M.M.P.; Manservigi, R. Replication-competent herpes simplex vectors: Design and applications. Gene Ther. 2005, 12, S170–S177. [Google Scholar] [CrossRef] [Green Version]
  160. Smith, T.R.F.; Patel, A.; Ramos, S.; Elwood, D.; Zhu, X.; Yan, J.; Gary, E.N.; Walker, S.N.; Schultheis, K.; Purwar, M.; et al. Immunogenicity of a DNA vaccine candidate for COVID-19. Nat. Commun. 2020, 11, 2601. [Google Scholar] [CrossRef]
  161. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines—A new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Vogel, A.B.; Lambert, L.; Kinnear, E.; Busse, D.; Erbar, S.; Reuter, K.C.; Wicke, L.; Perkovic, M.; Beissert, T.; Haas, H.; et al. Self-Amplifying RNA Vaccines Give Equivalent Protection against Influenza to mRNA Vaccines but at Much Lower Doses. Mol. Ther. 2018, 26, 446–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Walsh, E.E.; Frenck, R.W., Jr.; Falsey, A.R.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Neuzil, K.; Mulligan, M.J.; Bailey, R.; et al. Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine Candidates. N. Engl. J. Med. 2020, 383, 2439–2450. [Google Scholar] [CrossRef] [PubMed]
  164. Zeng, C.; Hou, X.; Yan, J.; Zhang, C.; Li, W.; Zhao, W.; Du, S.; Dong, Y. Leveraging mRNA Sequences and Nanoparticles to Deliver SARS-CoV-2 Antigens In Vivo. Adv. Mater. 2020, 32. [Google Scholar] [CrossRef]
  165. Shin, M.D.; Shukla, S.; Chung, Y.H.; Beiss, V.; Chan, S.K.; Ortega-Rivera, O.A.; Wirth, D.M.; Chen, A.; Sack, M.; Pokorski, J.K.; et al. COVID-19 vaccine development and a potential nanomaterial path forward. Nat. Nanotechnol. 2020, 15, 646–655. [Google Scholar] [CrossRef]
  166. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Marc, G.P.; Moreira, E.D.; Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
  167. Jackson, L.A.; Anderson, E.J.; Rouphael, N.G.; Roberts, P.C.; Makhene, M.; Coler, R.N.; McCullough, M.P.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; et al. An mRNA Vaccine against SARS-CoV-2—Preliminary Report. N. Engl. J. Med. 2020, 383, 1920–1931. [Google Scholar] [CrossRef]
  168. Anderson, E.J.; Rouphael, N.G.; Widge, A.T.; Jackson, L.A.; Roberts, P.C.; Makhene, M.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; Pruijssers, A.J.; et al. Safety and Immunogenicity of SARS-CoV-2 mRNA-1273 Vaccine in Older Adults. N. Engl. J. Med. 2020. [Google Scholar] [CrossRef]
  169. Lu, H. Drug treatment options for the 2019-new coronavirus (2019-nCoV). Biosci. Trends 2020, 14, 69–71. [Google Scholar] [CrossRef] [Green Version]
  170. Españo, E.; Kim, D.; Kim, J.; Park, S.-K.; Kim, J.-K. COVID-19 Antiviral and Treatment Candidates: Current Status. Immune Netw. 2021, 21. [Google Scholar] [CrossRef]
  171. Pujari, R.; Thommana, M.V.; Mercedes, B.R.; Serwat, A. Therapeutic Options for COVID-19: A Review. Cureus 2020, 12. [Google Scholar] [CrossRef] [PubMed]
  172. Mulangu, S.; Dodd, L.E.; Davey, R.T.; Tshiani Mbaya, O.; Proschan, M.; Mukadi, D.; Lusakibanza Manzo, M.; Nzolo, D.; Tshomba Oloma, A.; Ibanda, A.; et al. A Randomized, Controlled Trial of Ebola Virus Disease Therapeutics. N. Engl. J. Med. 2019, 381, 2293–2303. [Google Scholar] [CrossRef] [PubMed]
  173. Malin, J.J.; Suárez, I.; Priesner, V.; Fätkenheuer, G.; Rybniker, J. Remdesivir against COVID-19 and Other Viral Diseases. Clin. Microbiol. Rev. 2020, 34. [Google Scholar] [CrossRef]
  174. Siegel, D.; Hui, H.C.; Doerffler, E.; Clarke, M.O.; Chun, K.; Zhang, L.; Neville, S.; Carra, E.; Lew, W.; Ross, B.; et al. Discovery and Synthesis of a Phosphoramidate Prodrug of a Pyrrolo[2,1-f][triazin-4-amino] Adenine C-Nucleoside (GS-5734) for the Treatment of Ebola and Emerging Viruses. J. Med. Chem. 2017, 60, 1648–1661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Agostini, M.L.; Andres, E.L.; Sims, A.C.; Graham, R.L.; Sheahan, T.P.; Lu, X.; Smith, E.C.; Case, J.B.; Feng, J.Y.; Jordan, R.; et al. Coronavirus Susceptibility to the Antiviral Remdesivir (GS-5734) Is Mediated by the Viral Polymerase and the Proofreading Exoribonuclease. mBio 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  176. Tchesnokov, E.P.; Feng, J.Y.; Porter, D.P.; Götte, M. Mechanism of Inhibition of Ebola Virus RNA-Dependent RNA Polymerase by Remdesivir. Viruses 2019, 11, 326. [Google Scholar] [CrossRef] [Green Version]
  177. Beigel, J.H.; Tomashek, K.M.; Dodd, L.E.; Mehta, A.K.; Zingman, B.S.; Kalil, A.C.; Hohmann, E.; Chu, H.Y.; Luetkemeyer, A.; Kline, S.; et al. Remdesivir for the Treatment of Covid-19—Final Report. N. Engl. J. Med. 2020, 383, 1813–1826. [Google Scholar] [CrossRef]
  178. Furuta, Y.; Takahashi, K.; Kuno-Maekawa, M.; Sangawa, H.; Uehara, S.; Kozaki, K.; Nomura, N.; Egawa, H.; Shiraki, K. Mechanism of Action of T-705 against Influenza Virus. Antimicrob. Agents Chemother. 2005, 49, 981–986. [Google Scholar] [CrossRef] [Green Version]
  179. Delang, L.; Guerrero, N.S.; Tas, A.; Querat, G.; Pastorino, B.; Froeyen, M.; Dallmeier, K.; Jochmans, D.; Herdewijn, P.; Bello, F.; et al. Mutations in the chikungunya virus non-structural proteins cause resistance to favipiravir (T-705), a broad-spectrum antiviral. J. Antimicrob. Chemother. 2014, 69, 2770–2784. [Google Scholar] [CrossRef]
  180. Borrego, B.; de Ávila, A.I.; Domingo, E.; Brun, A. Lethal Mutagenesis of Rift Valley Fever Virus Induced by Favipiravir. Antimicrob. Agents Chemother. 2019, 63. [Google Scholar] [CrossRef] [Green Version]
  181. De Ávila, A.I.; Gallego, I.; Soria, M.E.; Gregori, J.; Quer, J.; Esteban, J.I.; Rice, C.M.; Domingo, E.; Perales, C. Lethal Mutagenesis of Hepatitis C Virus Induced by Favipiravir. PLoS ONE 2016, 11, e0164691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Escribano-Romero, E.; de Oya, N.J.; Domingo, E.; Saiz, J.C. Extinction of West Nile Virus by Favipiravir through Lethal Mutagenesis. Antimicrob. Agents Chemother. 2017, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Cai, Q.; Yang, M.; Liu, D.; Chen, J.; Shu, D.; Xia, J.; Liao, X.; Gu, Y.; Cai, Q.; Yang, Y.; et al. Experimental Treatment with Favipiravir for COVID-19: An Open-Label Control Study. Engineering 2020. [Google Scholar] [CrossRef]
  184. Doi, Y.; Hibino, M.; Hase, R.; Yamamoto, M.; Kasamatsu, Y.; Hirose, M.; Mutoh, Y.; Homma, Y.; Terada, M.; Ogawa, T.; et al. A Prospective, Randomized, Open-Label Trial of Early versus Late Favipiravir Therapy in Hospitalized Patients with COVID-19. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef]
  185. Corbett, A.H.; Lim, M.L.; Kashuba, A.D.M. Kaletra (Lopinavir/Ritonavir). Ann. Pharmacother. 2002, 36, 1193–1203. [Google Scholar] [CrossRef] [PubMed]
  186. Nutho, B.; Mahalapbutr, P.; Hengphasatporn, K.; Pattaranggoon, N.C.; Simanon, N.; Shigeta, Y.; Hannongbua, S.; Rungrotmongkol, T. Why Are Lopinavir and Ritonavir Effective against the Newly Emerged Coronavirus 2019? Atomistic Insights into the Inhibitory Mechanisms. Biochemistry 2020, 59, 1769–1779. [Google Scholar] [CrossRef] [Green Version]
  187. Wang, Z.; Chen, X.; Lu, Y.; Chen, F.; Zhang, W. Clinical characteristics and therapeutic procedure for four cases with 2019 novel coronavirus pneumonia receiving combined Chinese and Western medicine treatment. Biosci. Trends 2020, 14, 64–68. [Google Scholar] [CrossRef] [Green Version]
  188. Kim, J.Y. Letter to the Editor: Case of the Index Patient Who Caused Tertiary Transmission of Coronavirus Disease 2019 in Korea: The Application of Lopinavir/Ritonavir for the Treatment of COVID-19 Pneumonia Monitored by Quantitative RT-PCR. J. Korean Med. Sci. 2020, 35, e88. [Google Scholar] [CrossRef]
  189. Li, Y.; Xie, Z.; Lin, W.; Cai, W.; Wen, C.; Guan, Y.; Mo, X.; Wang, J.; Wang, Y.; Peng, P.; et al. Efficacy and Safety of Lopinavir/Ritonavir or Arbidol in Adult Patients with Mild/Moderate COVID-19: An Exploratory Randomized Controlled Trial. Medicine 2020, 1, 105–113.e4. [Google Scholar] [CrossRef]
  190. Horby, P.W.; Mafham, M.; Bell, J.L.; Linsell, L.; Staplin, N.; Emberson, J.; Palfreeman, A.; Raw, J.; Elmahi, E.; Prudon, B.; et al. Lopinavir–ritonavir in patients admitted to hospital with COVID-19 (RECOVERY): A randomised, controlled, open-label, platform trial. Lancet 2020, 396, 1345–1352. [Google Scholar] [CrossRef]
  191. WHO Solidarity Trial Consortium; Pan, H.; Peto, R.; Henao-Restrepo, A.-M.; Preziosi, M.-P.; Sathiyamoorthy, V.; Karim, Q.A.; Alejandria, M.M.; Garcia, C.H.; Kieny, M.P.; et al. WHO Solidarity Trial Consortium. Repurposed Antiviral Drugs for Covid-19—Interim WHO Solidarity Trial Results. N. Engl. J. Med. 2020, 384, 497–511. [Google Scholar] [CrossRef]
  192. Totura, A.L.; Bavari, S. Broad-spectrum coronavirus antiviral drug discovery. Expert Opin. Drug Discov. 2019, 14, 397–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Falzarano, D.; de Wit, E.; Rasmussen, A.; Feldmann, F.; Okumura, A.; Scott, D.P.; Brining, D.; Bushmaker, T.; Martellaro, C.; Baseler, L.; et al. Treatment with interferon-α2b and ribavirin improves outcome in MERS-CoV–infected rhesus macaques. Nat. Med. 2013, 19, 1313–1317. [Google Scholar] [CrossRef] [PubMed]
  194. Omrani, A.; Saad, M.M.; Baig, K.; Bahloul, A.; Abdul-Matin, M.; Alaidaroos, A.Y.; Almakhlafi, G.A.; Albarrak, M.M.; Memish, Z.A.; Albarrak, A.M. Ribavirin and interferon alfa-2a for severe Middle East respiratory syndrome coronavirus infection: A retrospective cohort study. Lancet Infect. Dis. 2014, 14, 1090–1095. [Google Scholar] [CrossRef] [Green Version]
  195. Arabi, Y.M.; Shalhoub, S.; Mandourah, Y.; Al-Hameed, F.; Al-Omari, A.; Al Qasim, E.; Jose, J.; Alraddadi, B.; AlMotairi, A.; Al Khatib, K.; et al. Ribavirin and Interferon Therapy for Critically Ill Patients with Middle East Respiratory Syndrome: A Multicenter Observational Study. Clin. Infect. Dis. 2020, 70, 1837–1844. [Google Scholar] [CrossRef] [Green Version]
  196. Hung, I.F.-N.; Lung, K.-C.; Tso, E.Y.-K.; Liu, R.; Chung, T.W.-H.; Chu, M.-Y.; Ng, Y.-Y.; Lo, J.; Chan, J.; Tam, A.R.; et al. Triple combination of interferon beta-1b, lopinavir–ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: An open-label, randomised, phase 2 trial. Lancet 2020, 395, 1695–1704. [Google Scholar] [CrossRef]
  197. Huang, Y.-Q.; Tang, S.-Q.; Xu, X.-L.; Zeng, Y.-M.; He, X.-Q.; Li, Y.; Harypursat, V.; Lu, Y.-Q.; Wan, Y.; Zhang, L.; et al. No Statistically Apparent Difference in Antiviral Effectiveness Observed Among Ribavirin Plus Interferon-Alpha, Lopinavir/Ritonavir Plus Interferon-Alpha, and Ribavirin Plus Lopinavir/Ritonavir Plus Interferon-Alpha in Patients with Mild to Moderate Coronavirus Disease 2019: Results of a Randomized, Open-Labeled Prospective Study. Front. Pharmacol. 2020, 11, 1071. [Google Scholar] [CrossRef] [PubMed]
  198. Kadam, R.U.; Wilson, I.A. Structural basis of influenza virus fusion inhibition by the antiviral drug Arbidol. Proc. Natl. Acad. Sci. USA 2017, 114, 206–214. [Google Scholar] [CrossRef] [Green Version]
  199. Wang, Z.; Yang, B.; Li, Q.; Wen, L.; Zhang, R. Clinical Features of 69 Cases with Coronavirus Disease 2019 in Wuhan, China. Clin. Infect. Dis. 2020, 71, 769–777. [Google Scholar] [CrossRef] [Green Version]
  200. Nojomi, M.; Yassin, Z.; Keyvani, H.; Makiani, M.J.; Roham, M.; Laali, A.; Dehghan, N.; Navaei, M.; Ranjbar, M. Effect of Arbidol (Umifenovir) on COVID-19: A randomized controlled trial. BMC Infect. Dis. 2020, 20, 1–10. [Google Scholar] [CrossRef] [PubMed]
  201. Lian, N.; Xie, H.; Lin, S.; Huang, J.; Zhao, J.; Lin, Q. Umifenovir treatment is not associated with improved outcomes in patients with coronavirus disease 2019: A retrospective study. Clin. Microbiol. Infect. 2020, 26, 917–921. [Google Scholar] [CrossRef]
  202. Hashem, A.M.; Alghamdi, B.S.; Algaissi, A.A.; Alshehri, F.S.; Bukhari, A.; Alfaleh, M.; Memish, Z.A. Therapeutic use of chloroquine and hydroxychloroquine in COVID-19 and other viral infections: A narrative review. Travel Med. Infect. Dis. 2020, 35, 101735. [Google Scholar] [CrossRef]
  203. Khan, M.; Santhosh, S.; Tiwari, M.; Rao, P.L.; Parida, M. Assessment of in vitro prophylactic and therapeutic efficacy of chloroquine against chikungunya virus in vero cells. J. Med. Virol. 2010, 82, 817–824. [Google Scholar] [CrossRef]
  204. Zhou, D.; Dai, S.-M.; Tong, Q. COVID-19: A recommendation to examine the effect of hydroxychloroquine in preventing infection and progression. J. Antimicrob. Chemother. 2020, 75, 1667–1670. [Google Scholar] [CrossRef] [PubMed]
  205. Retallack, H.; Di Lullo, E.; Arias, C.; Knopp, K.A.; Laurie, M.T.; Sandoval-Espinosa, C.; Leon, W.R.M.; Krencik, R.; Ullian, E.M.; Spatazza, J.; et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc. Natl. Acad. Sci. USA 2016, 113, 14408–14413. [Google Scholar] [CrossRef] [Green Version]
  206. Madrid, P.B.; Panchal, R.; Warren, T.K.; Shurtleff, A.C.; Endsley, A.N.; Green, C.E.; Kolokoltsov, A.; Davey, R.; Manger, I.D.; Gilfillan, L.; et al. Evaluation of Ebola Virus Inhibitors for Drug Repurposing. ACS Infect. Dis. 2015, 1, 317–326. [Google Scholar] [CrossRef] [PubMed]
  207. Bacharier, L.B.; Guilbert, T.W.; Mauger, D.T.; Boehmer, S.J.; Beigelman, A.; Fitzpatrick, A.M.; Jackson, D.J.; Baxi, S.N.; Benson, M.; Burnham, C.-A.D.; et al. Early Administration of Azithromycin and Prevention of Severe Lower Respiratory Tract Illnesses in Preschool Children with a History of Such Illnesses. JAMA 2015, 314, 2034–2044. [Google Scholar] [CrossRef]
  208. Gautret, P.; Lagier, J.C.; Parola, P.; Hoang, V.T.; Meddeb, L.; Mailhe, M.; Doudier, B.; Courjon, J.; Giordanengo, V.; Vieira, V.E.; et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: Results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents 2020, 56, 105949. [Google Scholar] [CrossRef]
  209. The RECOVERY Collaborative Group; Effect of Hydroxychloroquine in Hospitalized Patients with Covid-19. N. Engl. J. Med. 2020, 383, 2030–2040. [CrossRef] [PubMed]
  210. Bloch, E.M.; Shoham, S.; Casadevall, A.; Sachais, B.S.; Shaz, B.; Winters, J.L.; van Buskirk, C.; Grossman, B.J.; Joyner, M.; Henderson, J.P.; et al. Deployment of convalescent plasma for the prevention and treatment of COVID-19. J. Clin. Investig. 2020, 130, 2757–2765. [Google Scholar] [CrossRef] [Green Version]
  211. Erkurt, M.A.; Sarici, A.; Berber, I.; Kuku, I.; Kaya, E.; Özgül, M. Life-saving effect of convalescent plasma treatment in covid-19 disease: Clinical trial from eastern Anatolia. Transfus. Apher. Sci. 2020, 59, 102867. [Google Scholar] [CrossRef]
  212. Simonovich, V.A.; Pratx, L.D.B.; Scibona, P.; Beruto, M.V.; Vallone, M.G.; Vázquez, C.; Savoy, N.; Giunta, D.H.; Pérez, L.G.; Sánchez, M.D.L.; et al. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N. Engl. J. Med. 2020. [Google Scholar] [CrossRef]
  213. Barone, P.; DeSimone, R.A. Convalescent plasma to treat coronavirus disease 2019 (COVID-19): Considerations for clinical trial design. Transfusion 2020, 60, 1123–1127. [Google Scholar] [CrossRef]
  214. Wang, C.; Li, W.; Drabek, D.; Okba, N.M.A.; van Haperen, R.; Osterhaus, A.D.M.E.; van Kuppeveld, F.J.M.; Haagmans, B.L.; Grosveld, F.; Bosch, B.-J. A human monoclonal antibody blocking SARS-CoV-2 infection. Nat. Commun. 2020, 11, 1–6. [Google Scholar] [CrossRef] [PubMed]
  215. Wan, J.; Xing, S.; Ding, L.; Wang, Y.; Gu, C.; Wu, Y.; Rong, B.; Li, C.; Wang, S.; Chen, K.; et al. Human-IgG-Neutralizing Monoclonal Antibodies Block the SARS-CoV-2 Infection. Cell Rep. 2020, 32, 107918. [Google Scholar] [CrossRef] [PubMed]
  216. Wu, Y.; Wang, F.; Shen, C.; Peng, W.; Li, D.; Zhao, C.; Li, Z.; Li, S.; Bi, Y.; Yang, Y.; et al. A noncompeting pair of human neutralizing antibodies block COVID-19 virus binding to its receptor ACE2. Science 2020, 368, 1274–1278. [Google Scholar] [CrossRef] [PubMed]
  217. Brouwer, P.J.M.; Caniels, T.G.; van der Straten, K.; Snitselaar, J.L.; Aldon, Y.; Bangaru, S.; Torres, J.L.; Okba, N.M.A.; Claireaux, M.; Kerster, G.; et al. Potent neutralizing antibodies from COVID-19 patients define multiple targets of vulnerability. Science 2020, 369, 643–650. [Google Scholar] [CrossRef]
  218. Chen, P.; Nirula, A.; Heller, B.; Gottlieb, R.L.; Boscia, J.; Morris, J.; Huhn, G.; Cardona, J.; Mocherla, B.; Stosor, V.; et al. SARS-CoV-2 Neutralizing Antibody LY-CoV555 in Outpatients with Covid-19. N. Engl. J. Med. 2020. [Google Scholar] [CrossRef]
  219. Weinreich, D.M.; Sivapalasingam, S.; Perry, C.; Pan, C.; Hosain, R.; Mahmood, A.; Davis, J.D.; Turner, K.C.; Hooper, A.T.; Hamilton, J.D.; et al. REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19. N. Engl. J. Med. 2021, 384, 238–251. [Google Scholar] [CrossRef]
  220. Liu, B.; Li, M.; Zhou, Z.; Guan, X.; Xiang, Y. Can we use interleukin-6 (IL-6) blockade for coronavirus disease 2019 (COVID-19)-induced cytokine release syndrome (CRS)? J. Autoimmun. 2020, 111, 102452. [Google Scholar] [CrossRef] [PubMed]
  221. Zhou, M.; Zhang, X.; Qu, J. Coronavirus disease 2019 (COVID-19): A clinical update. Front. Med. 2020, 14, 126–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Le, R.Q.; Li, L.; Yuan, W.; Shord, S.S.; Nie, L.; Habtemariam, B.A.; Przepiorka, D.; Farrell, A.T.; Pazdur, R. FDA Approval Summary: Tocilizumab for Treatment of Chimeric Antigen Receptor T Cell-Induced Severe or Life-Threatening Cytokine Release Syndrome. Oncology 2018, 23, 943–947. [Google Scholar] [CrossRef] [Green Version]
  223. Guaraldi, G.; Meschiari, M.; Cozzi-Lepri, A.; Milic, J.; Tonelli, R.; Menozzi, M.; Franceschini, E.; Cuomo, G.; Orlando, G.; Borghi, V.; et al. Tocilizumab in patients with severe COVID-19: A retrospective cohort study. Lancet Rheumatol. 2020, 2, e474–e484. [Google Scholar] [CrossRef]
  224. Biran, N.; Ip, A.; Ahn, J.; Go, R.C.; Wang, S.; Mathura, S.; A Sinclaire, B.; Bednarz, U.; Marafelias, M.; Hansen, E.; et al. Tocilizumab among patients with COVID-19 in the intensive care unit: A multicentre observational study. Lancet Rheumatol. 2020, 2, e603–e612. [Google Scholar] [CrossRef]
  225. Xu, X.; Han, M.; Li, T.; Sun, W.; Wang, D.; Fu, B.; Zhou, Y.; Zheng, X.; Yang, Y.; Li, X.; et al. Effective treatment of severe COVID-19 patients with tocilizumab. Proc. Natl. Acad. Sci. USA 2020, 117, 10970–10975. [Google Scholar] [CrossRef]
  226. Klopfenstein, T.; Zayet, S.; Lohse, A.; Balblanc, J.-C.; Badie, J.; Royer, P.-Y.; Toko, L.; Mezher, C.; Kadiane-Oussou, N.J.; Bossert, M.; et al. Tocilizumab therapy reduced intensive care unit admissions and/or mortality in COVID-19 patients. Méd. Mal. Infect. 2020, 50, 397–400. [Google Scholar] [CrossRef] [PubMed]
  227. Strohbehn, G.W.; Heiss, B.L.; Rouhani, S.J.; Trujillo, J.A.; Yu, J.; Kacew, A.J.; Higgs, E.F.; Bloodworth, J.C.; Cabanov, A.; Wright, R.C.; et al. COVIDOSE: A Phase II Clinical Trial of Low-Dose Tocilizumab in the Treatment of Noncritical COVID-19 Pneumonia. Clin. Pharmacol. Ther. 2021, 109, 688–696. [Google Scholar] [CrossRef]
  228. Salama, C.; Han, J.; Yau, L.; Reiss, W.G.; Kramer, B.; Neidhart, J.D.; Criner, G.J.; Kaplan-Lewis, E.; Baden, R.; Pandit, L.; et al. Tocilizumab in Patients Hospitalized with Covid-19 Pneumonia. N. Engl. J. Med. 2021, 384, 20–30. [Google Scholar] [CrossRef] [PubMed]
  229. Stone, J.H.; Frigault, M.J.; Serling-Boyd, N.J.; Fernandes, A.D.; Harvey, L.; Foulkes, A.S.; Horick, N.K.; Healy, B.C.; Shah, R.; Bensaci, A.M.; et al. Efficacy of Tocilizumab in Patients Hospitalized with Covid-19. N. Engl. J. Med. 2020, 383, 2333–2344. [Google Scholar] [CrossRef]
  230. Tsai, A.; Diawara, O.; Nahass, R.G.; Brunetti, L. Impact of tocilizumab administration on mortality in severe COVID-19. Sci. Rep. 2020, 10, 1–7. [Google Scholar] [CrossRef]
  231. Leonard, W.J.; O’Shea, J.J. JAKS AND STATS: Biological Implications. Annu. Rev. Immunol. 1998, 16, 293–322. [Google Scholar] [CrossRef] [Green Version]
  232. Kontzias, A.; Kotlyar, A.; Laurence, A.; Changelian, P.; O’Shea, J.J. Jakinibs: A new class of kinase inhibitors in cancer and autoimmune disease. Curr. Opin. Pharmacol. 2012, 12, 464–470. [Google Scholar] [CrossRef] [Green Version]
  233. Bechman, K.; Subesinghe, S.; Norton, S.; Atzeni, F.; Galli, M.; Cope, A.P.; Winthrop, K.L.; Galloway, J.B. A systematic review and meta-analysis of infection risk with small molecule JAK inhibitors in rheumatoid arthritis. Rheumatology 2019, 58, 1755–1766. [Google Scholar] [CrossRef]
  234. 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, 395, e30–e31. [Google Scholar] [CrossRef] [Green Version]
  235. Cantini, F.; Niccoli, L.; Matarrese, D.; Nicastri, E.; Stobbione, P.; Goletti, D. Baricitinib therapy in COVID-19: A pilot study on safety and clinical impact. J. Infect. 2020, 81, 318–356. [Google Scholar] [CrossRef]
  236. Cantini, F.; Niccoli, L.; Nannini, C.; Matarrese, D.; Di Natale, M.E.; Lotti, P.; Aquilini, D.; Landini, G.; Cimolato, B.; Di Pietro, M.A.; et al. Beneficial impact of Baricitinib in COVID-19 moderate pneumonia; multicentre study. J. Infect. 2020, 81, 647–679. [Google Scholar] [CrossRef] [PubMed]
  237. Kalil, A.C.; Patterson, T.F.; Mehta, A.K.; Tomashek, K.M.; Wolfe, C.R.; Ghazaryan, V.; Marconi, V.C.; Ruiz-Palacios, G.M.; Hsieh, L.; Kline, S.; et al. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. N. Engl. J. Med. 2021, 384, 795–807. [Google Scholar] [CrossRef] [PubMed]
  238. Vaughan, C.J.; Gotto, A.M.; Basson, C.T. The evolving role of statins in the management of atherosclerosis. J. Am. Coll. Cardiol. 2000, 35, 1–10. [Google Scholar] [CrossRef] [Green Version]
  239. Vekic, J.; Zeljkovic, A.; Stefanovic, A.; Jelic-Ivanovic, Z.; Spasojevic-Kalimanovska, V. Obesity and dyslipidemia. Metabolism 2019, 92, 71–81. [Google Scholar] [CrossRef] [PubMed]
  240. Gorabi, A.M.; Kiaie, N.; Bianconi, V.; Jamialahmadi, T.; Al-Rasadi, K.; Johnston, T.P.; Pirro, M.; Sahebkar, A. Antiviral effects of statins. Prog. Lipid Res. 2020, 79, 101054. [Google Scholar] [CrossRef]
  241. Takahashi, T.; Suzuki, T. Function of Membrane Rafts in Viral Lifecycles and Host Cellular Response. Biochem. Res. Int. 2011, 2011, 1–23. [Google Scholar] [CrossRef] [Green Version]
  242. Fecchi, K.; Anticoli, S.; Peruzzu, D.; Iessi, E.; Gagliardi, M.C.; Matarrese, P.; Ruggieri, A. Coronavirus Interplay with Lipid Rafts and Autophagy Unveils Promising Therapeutic Targets. Front. Microbiol. 2020, 11, 1821. [Google Scholar] [CrossRef]
  243. Castiglione, V.; Chiriacò, M.; Emdin, M.; Taddei, S.; Vergaro, G. Statin therapy in COVID-19 infection. Eur. Hear. J. Cardiovasc. Pharmacother. 2020, 6, 258–259. [Google Scholar] [CrossRef] [PubMed]
  244. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef] [PubMed]
  245. Shin, Y.H.; Min, J.J.; Lee, J.-H.; Kim, E.-H.; Kim, G.E.; Kim, M.H.; Lee, J.J.; Ahn, H.J. The effect of fluvastatin on cardiac fibrosis and angiotensin-converting enzyme-2 expression in glucose-controlled diabetic rat hearts. Hear. Vessel. 2016, 32, 618–627. [Google Scholar] [CrossRef]
  246. Tikoo, K.; Patel, G.; Kumar, S.; Karpe, P.A.; Sanghavi, M.; Malek, V.; Srinivasan, K. Tissue specific up regulation of ACE2 in rabbit model of atherosclerosis by atorvastatin: Role of epigenetic histone modifications. Biochem. Pharmacol. 2015, 93, 343–351. [Google Scholar] [CrossRef]
  247. Wang, Q.; Zhang, Y.; Wu, L.; Niu, S.; Song, C.; Zhang, Z.; Lu, G.; Qiao, C.; Hu, Y.; Yuen, K.-Y.; et al. Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell 2020, 181, 894–904.e9. [Google Scholar] [CrossRef]
  248. Liao, J.K.; Laufs, U. Pleiotropic effects of statins. Annu. Rev. Pharmacol. Toxicol. 2005, 45, 89–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  249. Bifulco, M.; Gazzerro, P. Statin therapy in COVID-19 infection: Much more than a single pathway. Eur. Hear. J. Cardiovasc. Pharmacother. 2020, 6, 410–411. [Google Scholar] [CrossRef]
  250. Perucha, E.; Melchiotti, R.; A Bibby, J.; Wu, W.; Frederiksen, K.S.; Roberts, C.A.; Hall, Z.; LeFriec, G.; Robertson, K.A.; Lavender, P.; et al. The cholesterol biosynthesis pathway regulates IL-10 expression in human Th1 cells. Nat. Commun. 2019, 10, 1–13. [Google Scholar] [CrossRef] [Green Version]
  251. Zhang, X.-J.; Qin, J.-J.; Cheng, X.; Shen, L.; Zhao, Y.-C.; Yuan, Y.; Lei, F.; Chen, M.-M.; Yang, H.; Bai, L.; et al. In-Hospital Use of Statins Is Associated with a Reduced Risk of Mortality among Individuals with COVID-19. Cell Metab. 2020, 32, 176–187.e4. [Google Scholar] [CrossRef]
  252. Daniels, L.B.; Sitapati, A.M.; Zhang, J.; Zou, J.; Bui, Q.M.; Ren, J.; Longhurst, C.A.; Criqui, M.H.; Messer, K. Relation of Statin Use Prior to Admission to Severity and Recovery Among COVID-19 Inpatients. Am. J. Cardiol. 2020, 136, 149–155. [Google Scholar] [CrossRef] [PubMed]
  253. Mitacchione, G.; Schiavone, M.; Curnis, A.; Arca, M.; Antinori, S.; Gasperetti, A.; Mascioli, G.; Severino, P.; Sabato, F.; Caracciolo, M.M.; et al. Impact of prior statin use on clinical outcomes in COVID-19 patients: Data from tertiary referral hospitals during COVID-19 pandemic in Italy. J. Clin. Lipidol. 2021, 15, 68–78. [Google Scholar] [CrossRef]
  254. Levi, M.; Thachil, J.; Iba, T.; Levy, J.H. Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol. 2020, 7, e438–e440. [Google Scholar] [CrossRef]
  255. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef] [PubMed]
  256. Magro, C.; Mulvey, J.J.; Berlin, D.; Nuovo, G.; Salvatore, S.; Harp, J.; Baxter-Stoltzfus, A.; Laurence, J. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: A report of five cases. Transl. Res. 2020, 220, 1–13. [Google Scholar] [CrossRef] [PubMed]
  257. Tang, N.; Bai, H.; Chen, X.; Gong, J.; Li, D.; Sun, Z. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J. Thromb. Haemost. 2020, 18, 1094–1099. [Google Scholar] [CrossRef] [PubMed]
  258. Schiavone, M.; Gasperetti, A.; Mancone, M.; Curnis, A.; Mascioli, G.; Mitacchione, G.; Busana, M.; Sabato, F.; Gobbi, C.; Antinori, S.; et al. Oral anticoagulation and clinical outcomes in COVID-19: An Italian multicenter experience. Int. J. Cardiol. 2021, 323, 276–280. [Google Scholar] [CrossRef] [PubMed]
  259. Mycroft-West, C.J.; Su, D.; Pagani, I.; Rudd, T.R.; Elli, S.; Gandhi, N.S.; Guimond, S.E.; Miller, G.J.; Meneghetti, M.C.Z.; Nader, H.B.; et al. Heparin Inhibits Cellular Invasion by SARS-CoV-2: Structural Dependence of the Interaction of the Spike S1 Receptor-Binding Domain with Heparin. Thromb. Haemost. 2020, 120, 1700–1715. [Google Scholar] [CrossRef]
  260. Young, E. The anti-inflammatory effects of heparin and related compounds. Thromb. Res. 2008, 122, 743–752. [Google Scholar] [CrossRef]
  261. Drucker, D.J. Diabetes, obesity, metabolism, and SARS-CoV-2 infection: The end of the beginning. Cell Metab. 2021, 33, 479–498. [Google Scholar] [CrossRef]
  262. Bai, Y.; Lian, P.; Li, J.; Zhang, Z.; Qiao, J. The active GLP-1 analogue liraglutide alleviates H9N2 influenza virus-induced acute lung injury in mice. Microb. Pathog. 2020, 150, 104645. [Google Scholar] [CrossRef] [PubMed]
  263. Bloodworth, M.H.; Rusznak, M.; Pfister, C.C.; Zhang, J.; Bastarache, L.; Calvillo, S.A.; Chappell, J.D.; Boyd, K.L.; Toki, S.; Newcomb, D.C.; et al. Glucagon-like peptide 1 receptor signaling attenuates respiratory syncytial virus–induced type 2 responses and immunopathology. J. Allergy Clin. Immunol. 2018, 142, 683–687.e12. [Google Scholar] [CrossRef] [Green Version]
  264. Sato, T.; Shimizu, T.; Fujita, H.; Imai, Y.; Drucker, D.J.; Seino, Y.; Yamada, Y. GLP-1 Receptor Signaling Differentially Modifies the Outcomes of Sterile vs. Viral Pulmonary Inflammation in Male Mice. Endocrinology 2020, 161. [Google Scholar] [CrossRef]
  265. Alwarawrah, Y.; Nichols, A.G.; Green, W.D.; Eisner, W.; Kiernan, K.; Warren, J.; Hale, L.P.; Beck, M.A.; MacIver, N.J. Targeting T-cell oxidative metabolism to improve influenza survival in a mouse model of obesity. Int. J. Obes. 2020, 44, 2419–2429. [Google Scholar] [CrossRef] [PubMed]
  266. Chen, X.; Chen, Y.; Wu, C.; Wei, M.; Xu, J.; Chao, Y.-C.; Song, J.; Hou, D.; Zhang, Y.; Du, C.; et al. Coagulopathy is a major extrapulmonary risk factor for mortality in hospitalized patients with COVID-19 with type 2 diabetes. BMJ Open Diabetes Res. Care 2020, 8, e001851. [Google Scholar] [CrossRef] [PubMed]
  267. Schurink, B.; Roos, E.; Radonic, T.; Barbe, E.; Bouman, C.S.C.; de Boer, H.H.; de Bree, G.J.; Bulle, E.B.; Aronica, E.M.; Florquin, S.; et al. Viral presence and immunopathology in patients with lethal COVID-19: A prospective autopsy cohort study. Lancet Microbe 2020, 1, e290–e299. [Google Scholar] [CrossRef]
  268. Drucker, D.J. Coronavirus Infections and Type 2 Diabetes—Shared Pathways with Therapeutic Implications. Endocr. Rev. 2020, 41, 457–470. [Google Scholar] [CrossRef]
  269. Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [Green Version]
  270. Jiang, R.-D.; Liu, M.-Q.; Chen, Y.; Shan, C.; Zhou, Y.-W.; Shen, X.-R.; Li, Q.; Zhang, L.; Zhu, Y.; Si, H.-R.; et al. Pathogenesis of SARS-CoV-2 in Transgenic Mice Expressing Human Angiotensin-Converting Enzyme 2. Cell 2020, 182, 50–58.e8. [Google Scholar] [CrossRef]
  271. Bao, L.; Deng, W.; Huang, B.; Gao, H.; Liu, J.; Ren, L.; Wei, Q.; Yu, P.; Xu, Y.; Qi, F.; et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nat. Cell Biol. 2020, 583, 830–833. [Google Scholar] [CrossRef]
  272. Sun, S.-H.; Chen, Q.; Gu, H.-J.; Yang, G.; Wang, Y.-X.; Huang, X.-Y.; Liu, S.-S.; Zhang, N.-N.; Li, X.-F.; Xiong, R.; et al. A Mouse Model of SARS-CoV-2 Infection and Pathogenesis. Cell Host Microbe 2020, 28, 124–133.e4. [Google Scholar] [CrossRef] [PubMed]
  273. Dinnon, K.H., 3rd; Leist, S.R.; Schäfer, A.; Edwards, C.E.; Martinez, D.R.; Montgomery, S.A.; West, A.; Yount, B.L., Jr.; Hou, Y.J.; Adams, L.E.; et al. A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures. Nature 2020, 586, 560–566. [Google Scholar] [CrossRef] [PubMed]
  274. Leist, S.R.; Dinnon, K.H.; Schäfer, A.; Tse, L.V.; Okuda, K.; Hou, Y.J.; West, A.; Edwards, C.E.; Sanders, W.; Fritch, E.J.; et al. A Mouse-Adapted SARS-CoV-2 Induces Acute Lung Injury and Mortality in Standard Laboratory Mice. Cell 2020, 183, 1070–1085.e12. [Google Scholar] [CrossRef]
  275. Ingalls, A.M.; Dickie, M.M.; Snell, G.D. Obese, a new mutation in the house mouse. J. Hered. 1950, 41, 317–318. [Google Scholar] [CrossRef] [PubMed]
  276. Hummel, K.P.; Dickie, M.M.; Coleman, D.L. Diabetes, a New Mutation in the Mouse. Science 1966, 153, 1127–1128. [Google Scholar] [CrossRef]
  277. Surwit, R.S.; Kuhn, C.M.; Cochrane, C.; McCubbin, J.A.; Feinglos, M.N. Diet-Induced Type II Diabetes in C57BL/6J Mice. Diabetes 1988, 37, 1163–1167. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of how age-related alterations escalate susceptibility of COVID-19. The aging immune system are subject to innate and adaptive immunosenescence, and inflammaging. An age-associated decrease in NAD+ leads to de-repression of NLRP3 and activation of inflammasome in older people, further aggravating the cytokine storm. The glycome regulating a variety of immune signaling pathways changes during aging.
Figure 1. Schematic diagram of how age-related alterations escalate susceptibility of COVID-19. The aging immune system are subject to innate and adaptive immunosenescence, and inflammaging. An age-associated decrease in NAD+ leads to de-repression of NLRP3 and activation of inflammasome in older people, further aggravating the cytokine storm. The glycome regulating a variety of immune signaling pathways changes during aging.
Cells 10 01412 g001
Figure 2. Schematic illustration of how obesity influence the development of COVID-19. Multiple pathological features of obesity, such as systematic chronic inflammation and dysfunctionality in ASCs/MSCs, immune response, metabolism, and endothelium, affect the progression and outcomes of COVID-19.
Figure 2. Schematic illustration of how obesity influence the development of COVID-19. Multiple pathological features of obesity, such as systematic chronic inflammation and dysfunctionality in ASCs/MSCs, immune response, metabolism, and endothelium, affect the progression and outcomes of COVID-19.
Cells 10 01412 g002
Table 1. COVID-19 vaccines currently in clinical trials or authorized.
Table 1. COVID-19 vaccines currently in clinical trials or authorized.
Company/OrganizationBrand NameVaccine Type/PlatformClinical Trial Status
Inactivated/Killed Vaccines
Bharat Biotech, Indian Council of Medical Research, National Institute of Virology, Ocugen, Precisa MedicamentosCovaxinInactivatedIII
Institute of Medical Biology, Chinese Academy of Medical SciencesCOVID-19 vaccineInactivated III
Sinovac, Instituto Butantan, Bio FarmaCoronaVac (PiCoVacc)Inactivated III
Beijing Institute of Biological Products, SinopharmBBIBP-CorVInactivated III
Wuhan Institute of Biological Products, SinopharmCOVID-19 vaccineInactivatedIII
Research Institute for Biological Safety Problems, Republic of KazakhstanQazCovid-InInactivatedIII
Non-Replicating or Replicating Viral Vector Vaccines
CanSino Biologics, Beijing Institute of Biotechnology, PetrovaxAd5-nCoV (Convidencia)Non-replicating viral vectorIII
AstraZeneca, University of Oxford, Serum Institute of IndiaChAdOx1 nCoV-19 (AZD1222)Non-replicating viral vector Authorized
Gameleya Research InstituteSputnik V (Gam-COVID-Vac)Non-replicating viral vector III
Janssen Pharmaceutical Companies of Johnson & JohnsonAd26.COV2.S (JNJ-78436725)Non-replicating viral vectorAuthorized
DNA-Based Vaccines
AnGes, Osaka University, Takara BioAG0301&AG0302Plasmid II/III
Inovio Pharmaceuticals, International Vaccine InstituteINO-4800Plasmid II/III
Zydus CadilaZyCov-DPlasmid III
RNA-Based Vaccines
CureVacCVnCoVLNP-mRNAIII
Moderna, NIAID (VRC)mRNA-1273LNP-mRNAAuthorized
Pfizer, BioNTech, Fosun PharmaBNT162b2LNP-mRNAAuthorized
Protein Subunit Vaccines
NovavaxNVX-CoV2373Full length recombinant SARS-COV-2 glycoprotein nanoparticle vaccine adjuvanted with Matrix MIII
Anhui Zhifei Longcom Biopharmaceutical, Chinese Academy of SciencesZF2001Adjuvanted recombinant protein (RBD-Dimer)III
Instituto Finlay de VacunasFINLAY-FR-2rRBD produced in CHO-cell chemically conjugate to tetanus toxoidIII
Table 2. Antiviral and therapeutic drugs for treating COVID-19.
Table 2. Antiviral and therapeutic drugs for treating COVID-19.
DrugTarget/Mode of ActionStatus
Remdesivir (RDV)Adenosine analog viral RdRp inhibitorEffects at middle COVID-19 stage demonstrated; Approved by the US-FDA
FavipiravirInfluenza virus emergency drug (Japan)
Purine analogViral RdRp inhibitor
Effects at early COVID-19 stage demonstrated; EUA (outside USA)
Lopinavir + ritonavirHIV protease inhibitor (approved)
Potential CoV protease inhibitor
Effects at early COVID-19 stage indicated
RibavirinGuanosine analog
Inhibits GTP synthesis Viral mutagenesis Immunomodulatory activity
Effects at early COVID-19 stage indicated
UmifenovirInfluenza treatment and prophylaxis (China and Japan)
Viral endocytosis inhibitor
Inhibitor of viral genome replication
Effects at early COVID-19 stage indicated
Chloroquine (CQ), hydroxychloroquine (HCQ)Anti-malaria
Anti-rheumatoid arthritis (HCQ)
Viral endocytosis inhibitor in vitro
Effects at early COVID-19 stage indicated
NAbs: LY-CoV555, REGN-COV2Viral neutralizationEffects at early COVID-19 stage demonstrated; EUA (USA)
TocilizumabIL-6 receptorEffects at middle to late COVID-19 stage demonstrated
BaricitinibJAK1/2Effects at middle to late COVID-19 stage indicated
Early COVID-19 stage, first week of infection, viral phase, pre-/early symptomatic phase; middle COVID-9 stage, second week of infection, symptomatic, early stages of hyperinflammation; late COVID-19 stage, beyond second week of infection, hyperinflammatory to thrombotic stages. EUA: Emergency use authorization.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Shin, C.-H.; Kim, K.-H.; Jeeva, S.; Kang, S.-M. Towards Goals to Refine Prophylactic and Therapeutic Strategies Against COVID-19 Linked to Aging and Metabolic Syndrome. Cells 2021, 10, 1412. https://0-doi-org.brum.beds.ac.uk/10.3390/cells10061412

AMA Style

Shin C-H, Kim K-H, Jeeva S, Kang S-M. Towards Goals to Refine Prophylactic and Therapeutic Strategies Against COVID-19 Linked to Aging and Metabolic Syndrome. Cells. 2021; 10(6):1412. https://0-doi-org.brum.beds.ac.uk/10.3390/cells10061412

Chicago/Turabian Style

Shin, Chong-Hyun, Ki-Hye Kim, Subbiah Jeeva, and Sang-Moo Kang. 2021. "Towards Goals to Refine Prophylactic and Therapeutic Strategies Against COVID-19 Linked to Aging and Metabolic Syndrome" Cells 10, no. 6: 1412. https://0-doi-org.brum.beds.ac.uk/10.3390/cells10061412

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop