explicitClick to confirm you are 18+

Therapeutic Modalities and Novel Approaches in Regenerative Medicine for COVID-19

MacKenzieOct 27, 2020, 5:40:19 AM
thumb_up2thumb_downmore_vert

https://www.sciencedirect.com/science/article/pii/S0924857920304192

Author links open overlay panelRoyaRamezankhaniabcd1RoyaSolhibe1ArashMemarnejadianfFatemeharefehNamicSeyed MohammadRezaHashemiangTineTricotcMassoudVosoughabCatherineVerfailliec

Show more

https://doi.org/10.1016/j.ijantimicag.2020.106208Get rights and content

Highlights

 

Direct cytopathic effect and immunopathological pathogenesis are as two main underlying mechanisms for severe pulmonary injury in COVID-19 infections.

Repurposed antiviral agents are among the most promising therapeutics for overcoming the COVID-19 pandemic.

Immunomodulatory effects of mesenchymal stem/stromal cells (MSCs) hold the potential for preventing the immune mediated consequences of SARS-CoV-2.

Nucleic acid- based approaches have gained much attention for treating COVID-19 infections.

 

Abstract

The recent coronavirus disease 2019 (COVID-19) outbreak around the world has an enormous impact on the global health burden, threatening the lives of many individuals, and has severe socio-economic consequences. Many pharmaceutical and biotechnology companies have begun intensive research on different therapeutic strategies, from repurposed antiviral drugs to vaccines and monoclonal antibodies to prevent the spread of the disease and treat infected patients. Among the various strategies, advanced therapeutic approaches including cell- and gene editing-based therapeutics are also being investigated and initial results in in vitro and early phase I studies were promising, however, still further assessments are required. In this paper, we first review the underlying mechanisms for severe acute respiratory syndrome coronavirus (SARS-CoV-2) pathogenesis and further discuss available therapeutic candidates and advanced modalities that are being currently evaluated in in vitro/in vivo models and of note in clinical trials.

Keywords

SARS-CoV-2

COVID-19

Advanced therapeutic approaches

ATMP

1. SARS-CoV-2 background

The recent outbreak of COVID-19, caused by a novel beta-coronavirus (CoV), is now a major worldwide medical (and economical) challenge. Therefore, specifying the therapeutic approaches and the mechanisms which lead to these strategies are of outmost importance. Reviewing the published papers in regards with the mechanisms and the state of art medications, we have tried to draw an overall picture of the involved mechanism and the related therapeutic approaches. The type of documents used to obtain the data were original articles, review articles, and HTML documents from the official websites (e.g. WHO). Search terms included MeSH (Medical Subject Headings) terms, “coronavirus, severe acute respiratory syndrome coronavirus 2, 2019-nCoV, along with focusing on novel therapeutic approaches”. The registered and active clinical trials were found on ClinicalTrials.gov and the index of studies of novel coronavirus pneumonia in the Chinese Clinical Trial Registry. The cut-off date for the data search was “September 2020.

CoVs are enveloped viruses, wherein the 27-32kb genomic RNA is capped and polyadenylated (1). They are subdivided into four distinct groups; alpha, beta, delta and gamma (2). The CoV species HKU1, NL63, OC43 and 229E cause common cold symptoms. Others, like the Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Coronavirus (MERS-CoV) result in fatal diseases. SARS-CoV-2 is the third virus in the CoV family with the potential to cause life-threatening disease in a wide range of individuals (3).

An Approximately 30 kb single‐stranded positive‐sense RNA (+ssRNA) forms the genomic content of SARS-CoV-2, and makes it the largest known RNA virus (4). The genome interacts with the nucleoprotein, N, which is bound to the membrane (M) protein. The M protein plays a crucial role in viral assembly and budding. The envelop (E) protein functions in viral morphogenesis, release and pathogenesis (5). Finally, the spike (S) protein is a trimeric glycoprotein including two subunits of S1 and S2 (Figure 1). Using the S1 and S2 subunits of S glycoprotein, coronaviruses have acquired the ability to attach and fuse to the target cell membrane, respectively (Figure 2b) (6). M, E, and S make up the virus envelop (5). The genome and sub-genome of a typical CoV contain at least six ORFs (4). Two-thirds of that include ORF1a, and 1b, which are translated into the polyproteins 1a (pp1a) and pp1ab, respectively (Figure 3a). The remaining ORFs on the one-third of the genome encode for the main structural [S, E, M, and N] and accessory proteins. After translation, the viral genome initiates replication. Most of the 16 nonstructural proteins (nsps), which are produced from pp1a and pp1ab form a very large protein complex, responsible for viral genome replication and subgenomic mRNA (sgmRNA) synthesis (7). The viral life cycle is completed by fusion of virus particles with the plasma membrane and release into the extracellular space. While MERS-CoV S protein binds to the dipeptidyl peptidase 4 (DPP4) receptor, to gain entry into the cells, the receptor for both SARS-CoV and SARS-CoV-2 is hACE2 (8).

Figure 1
  1. Download : Download high-res image (395KB)
  2. Download : Download full-size image

Figure 1. An overview of SARS-CoV-2 structure.

Figure 2
  1. Download : Download high-res image (929KB)
  2. Download : Download full-size image

Figure 2. Pathogenesis of SARS-CoV-2. A. fever and coughing along with headache are the common symptoms of COVID-19. ARDS along with multi organ failure account for the critical stage of the disease (Left). Type II alveolar cells highly express ACE2, the known receptor for SARS-CoV-2 (Right). B. Virus entry mechanism along with the involved enzymes in this process is shown.

Figure 3
  1. Download : Download high-res image (972KB)
  2. Download : Download full-size image

Figure 3. A. Viral genome replication and subgenomic mRNA (sgmRNA) synthesis represented. B. Interaction of APCs with lymphocytes and cytokine activation (Created by Biorender.com).

2. Pathogenesis

Aside from the oral mucosa, especially the tongue, high expression levels of the SARS-CoV-2 receptor, hACE2, have been reported on lung type II alveolar cells (AT2), upper and stratified esophageal epithelial cells, absorptive enterocytes from ileum and colon, cholangiocytes, myocardial cells, kidney proximal tubule cells, podocytes, bladder urothelial cells, male reproductive cells, placental trophoblasts, eye, and vascular endothelial cells (9, 10, 11). Two different pathophysiological patterns may be responsible for severe pulmonary injury in COVID-19 infections, direct cytopathic effect (CPE) and immunopathological pathogenesis. The cytopathic effect may be related to a high level of viral load, while the role of immune mediated pulmonary effects are more prevalent in late respiratory failure where the viral load has already been reduced (12). Both of these events are discussed in the following paragraphs.

2.1. Direct cytopathic effect

The exact mechanism(s) underlying the CPE of SARS-CoV-2 are not yet completely understood. However, as SARS-CoV-2 has high similarities with SARS-CoV (structure and entry-receptor specificity) as well as MERS-CoV (structure, but not entry-receptor), insights from how SARS-CoV and MERS-CoV cause pulmonary cell will undoubtedly speed the discovery of SARS-CoV-2 mediated pulmonary cell death (13). SARS-CoV causes cell death via both apoptosis and necrosis. Also, MERS-CoV has been shown to induce apoptosis in both immune and non-immune cells such as lung and kidney cells (14, 15). These findings can form the basis for possible underlying mechanisms through which SARS-CoV-2 may prompt its CPE, which has been demonstrated in human airway epithelial cells following the virus inoculation along with the cessation in cilia movements (3). Regarding the kidney cells there is evidence for a direct CPE of SARS-CoV-2 on various renal cells (16). For other cell types with ACE2 receptors, further studies need to be done to determine the probable direct CPE of SARS-CoV-2, as, for instance, in one study no apparent histological alterations in heart tissue was reported in the postmortem histopathological study on a COVID-19 infected patient other studies have suggested cardiac injury due to the direct effect of virus entry into myocardial tissue (17, 18, 19). Also, very little is known regarding the CPE of SARS-Cov-2 in gastrointestinal (GI) cells (20). Nevertheless, the increased rate of CPE after inoculation of human intestinal organoids and liver organoids with SARS-Cov-2 has been seen (21, 22). ACE2 is not expressed on hematopoietic cells; hence, direct infection of immune cells by SARS-CoV-2 may not be likely. However, if SARS-CoV-2 can infect the immune cells directly still needs to be studied (23).

2.2. Immunological effects

The precise mechanisms through which SARS-CoV-2 affects the immune system are not yet fully known. Similarities in the pathogenesis of coronaviruses, especially between SARS-CoV and SARS-CoV-2, have, however, provided valuable insights in how SARS-CoV-2 might affect the immune system (24).

Both innate and adaptive immune responses are involved in COVID-19 pathogenesis. Several components of the innate immune system have been reported to be over activated or increased in number. In fact, macrophage activation syndrome (MAS) is suggested as one of the possible reasons of COVID-19-related hyperinflammation (25). It is because SARS-CoV-2 has been shown to cause the activation of NLRP3 inflammasome in macrophages, which leads to an increased level of proinflammatory cytokines production (26). Moreover, neutrophils have also been predominantly found in lung infiltration of COVID-19 patients. The elevated amount of neutrophils and neutrophil-to-lymphocyte ratio (NLR) usually predict poor clinical outcome (27). In addition, it is shown that necroinflammation is one of the results of neutrophils infiltration and neutrophil extracellular traps (NETs) formation in COVID-19 patients (28). On the other hand, recognition of the pathogen-associated molecular pattern (PAMP) of SARS-CoV (genomic RNA) by TLR3 and TLR7 and the cytosolic RNA sensor, RIG-I/MDA5 subsequently lead to pro-inflammatory cytokine induction, particularly, type I IFN. However, both structural and non-structural proteins from coronaviruses interfere with the type I IFN-related signaling pathways (9, 29). The delayed type I IFN response along with the confirmed increase in neutrophils and monocytes/macrophages influx (30), followed by an excessive production of type I IFN in the later phases may explain part of the symptoms of the virus.

In addition to innate immunity, both humoral and cellular immune responses also play significant roles in coronavirus clinical complications. Many attempts to identify T and B cells epitopes for the virus structural proteins have been undertaken (31). Cytotoxic CD8 T-cells (cytotoxic T cells) and helper CD4 T-cells (helper T cells), as key parts of antiviral immunity, require the presentation of viral antigens through HLA I and II molecules on the surface of antigen presenting cells (APCs). Genetic polymorphisms in components of the antigen presentation system appear to account at least in part for the risk of SARS-CoV infections (29). In this regards, one in silico study showed that the expression of HLA-B*46:01 may make the individuals more susceptible to COVID-19 as the binding peptides for SARS-CoV-2 is predicted to be the least, while HLA-B*15:03 expression may enable cross-protective T-cell-based immunity (32). However, the correlation between different allele frequencies and susceptibility to SARS-CoV-2 infection needs more investigation. CD8+ T cells can destroy virally infected cells. CD4+ cells have a key role in promoting the activation of T‐dependent B cells and the production of proinflammatory cytokines leading to recruitment of monocytes and macrophages and over production of cytokine and chemokines (33). Reduction of T helper cells therefore, might lead to a strong immune‐mediated interstitial pneumonitis and delayed clearance of SARS‐CoV from lungs (34). It is also noteworthy that all the SARS‐CoV-related memory T cells found in SARS‐CoV convalescent patients, mediated an anti-SARS‐CoV structural protein response, whereby the S protein was mostly involved in these T cell responses (35, 36). This implies a role of the structural proteins as candidate for designing efficient SARS vaccines. Furthermore, clinical observations in COVID-19 patients confirm the reduction of excessively activated CD4+ and CD8+ T cells (18). Production of early stage-related IgM, long lasting specific IgG, and IgA forms the main B cell immune response to SARS-CoV (29). In this regards, the isolation of specific B-cell clones that produce neutralizing monoclonal antibodies has been already shown in a SARS-CoV convalescent patient (37). Although neutralizing antibodies may have the potential to block the viruses entry into human cells (38), anti-S protein neutralizing IgGs might also hold the risk for fatal acute lung injury (39).

On the other hand, it is interesting to note that ACE2 through which SARS-CoV-2 enters into cells has been reported to be downregulated in one mouse model of SARS-CoV infection and pulmonary disease, and that this downregulation may lead to more severe lung injuries (40). In the renin–angiotensin–aldosterone system (RAAS), ACE converts angiotensin I to angiotensin II, while ACE2 has a role in angiotensin II inactivation (40). Therefore, it is possible that SARS-CoV-2 can cause an increase in blood flow not only by the acute inflammation response, but also via an increase in angiotensin II due to ACE2 downregulation. In addition, the circulatory fraction of immunosenescent T cells (i.e. CD8+CD28−CD57+ cells that accumulate with aging and in chronic inflammatory conditions (41)) have been found in larger numbers of patients with high blood pressure, which may raise the risk for severe forms of COVID-19. This together with elevated levels of C-X-C chemokine receptor type 3 (CXCR3) chemokines and serum granzyme B, also hypothesized to be involved in T-cell–driven inflammation in human hypertension (42), may explain why aging and / or hypertension worsen the incidence and prognosis of COVID-19.

2.3. Cytokine storm-based effects

Acute respiratory distress syndrome (ARDS), a common immunopathological process, might be the leading cause of death in COVID-19 patients (Figure 3b) (43). In addition, failure of several other organs has been reported in severe cases of COVID-19 patients (18). The uncontrolled anti-COVID-19 proinflammatory cytokine and chemokine production, also termed cytokine storm, causes ARDS (44).

However, it is important to note that there are doubts regarding the relevancy of this event to COVID-19. In this regards, Sinha et al. have questioned the precise function of imbalanced cytokine responses in COVID-19 patients. They suggest that lung injury in COVID-19 patients is not solely attributed to “cytokine storm”. Of note, they mention that although the level of IL-6, a key cytokine in acute inflammation, in patients with COVID-19 is higher than the median value, it is less than what is measured in individuals affected by ARDS (10-200 fold higher in ARDS). Based on some available evidences, they note that, alveolar micro thrombi might be the possible culprits for lung injury in COVID-19 (45). Moreover, there are evidences indicating that the defect in both innate and adaptive immune responses is of more importance in comparison with the hypercytokinemia-induced organ injury, regarding the pathophysiological abnormalities in COVID -19 patients (46).

2.4. Coagulation dysfunctions

High blood pressure, pulmonary embolism and thrombosis are among the symptoms seen in COVID-19 patients, proposing the hypothesis that COVID-19 is an endothelial dysfunction disease (47, 48). In fact, significant higher levels of D-dimer and fibrin degradation products (FDP) together with longer prothrombin time (PT) has been confirmed in survivors compared to non-survivors upon admission to hospital (49). Coagulation dysfunction is related to the imbalanced immune response and massive inflammatory reactions, leading to microvascular system damage and activation of coagulation processes (50). This in turn leads to extensive microthrombosis (51). Due to the widespread inflammation, negative control mechanisms by which the thrombin production is monitored can be inhibited (51). The inflammatory reactions caused by the overproduction of proinflammatory cytokines also promote vascular permeability (51). On the other hand, reduced activity of ACE2 in lungs of animal models with coronavirus-induced severe ARDS may increase the risk of vascular hyperpermeability and pulmonary edema, as ACE2 is a negative regulatory factor for severity of lung edema (52). Pulmonary embolus is shown to be frequent in COVID-19 patients (53).

2.5. Circulating ACE2

ADAM17 and TMPRSS2 proteases are responsible for the cleavage of ACE2, which is mostly anchored at the apical surface of the cell (Figure 2b). However, while the first may have protective roles against SARS-CoV-2, the latter facilitates the virus entry (54, 55). In fact, metalloprotease ADAM17 cleaves the N-terminal catalytic domain of ACE2, which is also the coronavirus-binding site, and releases it into circulation. The exact role of cleaved ACE2 in circulation of COVID-19 patients still needs to become clear, however it was previously showed that serum ACE2 activity is elevated during hypertension and progression of cardiovascular disease (56, 57).

3. Therapeutics

Currently, the main strategy for managing the disease, focuses on supportive treatments such as oxygen therapy, fluid management, and ventilator support. However, it is of note to mention that despite the controversy on using the noninvasive ventilation (NIV) for managing ARDS in COVID-19 patients, still there might be a selected subpopulation of patients who may benefit from NIV (58).

Disease specific therapies might include available anti-viral medications as well as advanced strategies, such as cellular and gene-based protocols. In the following sections, we classified some of the currently suggested and available therapeutics based on the stated order for the virus pathogenesis mechanism (i.e. 1- Direct CPE, 2- Immunological effects and 3- Coagulation dysfunction) for management of COVID-19 patients (Figure 4). The defined therapeutics involves repurposed antiviral agents, cell/gene-based strategies, anti-cytokine modalities, and the anti-coagulation therapies.

Figure 4
  1. Download : Download high-res image (442KB)
  2. Download : Download full-size image

Figure 4. Therapeutics. A. The current therapeutics used for treating COVID-19 patients. B. Advanced therapeutic medicinal products (ATMP). Cell- and gene-based therapies that might be candidate therapies (Created by Biorender.com).

4. Prevention of direct cell death due to virus

4.1. Repurposed antiviral agents

Several existing antiviral drugs have been repurposed and used to treat COVID-19 infected patients. Remdesivir (RDV), an adenosine analogue, was originally developed for the treatment of Ebola virus disease. RDV may have antiviral activity against a number of other RNA viruses, including SARS–CoV and MERS–CoV (59, 60). The triphosphate form of RDV competes with adenosine triphosphate (ATP) for incorporation into the genome and inhibits the RNA-dependent RNA polymerase (59). Following promising results of the drug in vitro and in vivo studies against MERS-CoV and SARS-CoV (61), RDV is a FDA- approved emergency use authorized anti-COVID-19 drug which has been tested in multi-site clinical trials. Initial studies suggest that treating COVID-19 patients with intravenous RDV improves the clinical condition of patients, even if side effects such as temporary gastrointestinal upset have been noted (62). Recently, Beigel et al. evaluated beneficial effects of RDV in hospitalized COVID-19 patients. Based on their findings, RDV treatment can shorten the recovery time in patients (63).

Other nucleotide analogues such as favipiravir (nucleoside analog), ribavirin (guanosine analog), galidesivir (adenosine analog), sofosbuvir (pyrimidine nucleotide analogue), alovudine (thymidine dideoxynucleoside analogue), Zidovudine (ZDV) (thymidine analogue), etc. are also under investigation for treatment of COVID-19 (29, 64). Favipiravir is an RNA polymerase inhibitor for a number of RNA viruses, and is approved for treatment of influenza in China and Japan. Several clinical trials are investigating the therapeutic effect of favipiravir for COVID-19 (65). Favipiravir may have a more potent antiviral action than some protease inhibitors such as kaletra (lopinavir/ritonavir) (65). However, compared to arbidol (an inhibitor of membrane fusion between the virus and the plasma membrane and also endocytic vesicle membranes), no significant improvement was reported in clinical recovery rate of patients after 7 days of favipiravir therapy (66). It is of note that recently a small phase 3 trial in India with 150 patients showed faster viral clearance in mild to moderate COVID-19 cases who had received Favipiravir (67). Accordingly, favipiravir is approved for restricted emergency use in moderate COVID-19 cases by Drugs Controller General of India (DCGI) (68).

Other antiviral drugs such as oral oseltamivir (a neuraminidase inhibitor), intravenous ganciclovir, and chloroquine phosphate tablets are candidate drugs that may reduce the infection symptoms (69). At the beginning of the pandemic, there were some reports indicating that chloroquine phosphate (an anti-malarial agent), that has both anti-viral and anti-inflammatory activities, might prevent worsening of pneumonia. However, due to the serious cardiac adverse events and other potential serious side effects of chloroquine phosphate and hydroxychloroquine sulfate and the low efficacy against COVID-19, the U.S. Food and Drug Administration (FDA) has recently cancelled its emergency use authorization (EUA) (69, 70). Also, the combination of hydroxychloroquine and azithromycin has recently been shown to have no significant effect on the rate of virologic clearance in patients with COVID-19 (71). Consistently, Cavalcanti et al. observed that using hydroxychloroquine, alone or with azithromycin, has no effects in the clinical status of patients with COVID-19 (72).

Chymotrypsin-like proteases (3CLpro), such as cinanserin, and flavonoids along with papain-like proteases (PLP) like diarylheptanoids may prevent coronavirus replication and are also considered as candidates to battle the virus (29). Finally, low-dose systemic administration of corticosteroids in addition to inhalation of interferon are other anti-COVID-19 strategies (69). Recently, it has been shown that dexamethasone appears promising in reducing the mortality rate in critically ill COVID-19 patients, which is appreciated by WHO (73). In fact, it is shown that the use of dexamethasone (at a dose of 6 mg once daily) in patients hospitalized with COVID-19 leads to lower mortality rate (74).

As mentioned in the pathogenesis section, ACE2 is a receptor responsible for entry of the virus. On the other hand, S protein and TMPRSS2 are among the molecules that are essential for viral entry. By using neutralizing antibodies against S protein and also TMPRSS2 inhibitors (camostate mesylate), viral entry was blocked (75). Moreover, recombinant ACE2 (APN01) could reduce both angiotensin ІІ and IL-6 levels in ARDS patients. This agent is currently under investigation for COVID-19 patients in China (76). However, there is a challenge regarding the use of ACE inhibitors in the context of cardiovascular diseases. For example, high levels of urinary ACE2 has been detected in patients who received the angiotensin-receptor blocker Olmesartan, but this was not seen in patients receiving the ACE inhibitor, enalapril or other angiotensin-receptor blocker, losarthan (77). Thus, use of angiotensin-receptor blockers or ACE inhibitors in the treatment of COVID-19 needs further investigation (78).

4.2. mAbs

Researchers tested to neutralize SARS-CoV-2 with mAbs previously shown to bind to SARS-CoV receptor binding domain (RBD)-directed mAbs. However, no significant binding to SARS-CoV-2 was seen (79). On the other hand, SARS-specific human monoclonal antibody CR3022 may have some cross-reactive binding between SARS-CoV-2 and SARS-CoV (80). Therefore, investigating the cross reactivity of other mAbs against SARS-CoV, including m396 and CR3014, may have the potential for the treatment of COVID-19 patients (29).

Moreover, neutralizing antibodies in the plasma of patients recovering from SARS, MERS, or the 2009 H1N1 pandemic, could modify the disease progression of other patients with these infections. Therefore, applying convalescent plasma (CP) from COVID-19 patients might hold promise to attenuate the clinical symptoms and mitigating the pulmonary damage, and eliminate SARS-CoV-2 RNA clearance (81, 82). In this regards, FDA has issued an EUA for COVID-19 convalescent plasma, although the significant efficacy of this approach for the treatment of COVID-19 patients is yet to be demonstrated in placebo-controlled randomized controlled trials (RCTs) (83).

4.3. Gene therapy-based strategies

In the context of development of novel treatments for viral infections including SARS-CoV-2, nucleic acid- based strategies as well as clustered regularly interspaced short palindromic repeats (CRISPR) associated protein nuclease (Cas)-based approaches could be used.

4.3.1. siRNA

The potential of siRNA technology for treating viral infections have been previously shown (84). Therefore, siRNAs may be considered as potential candidates to be used against SARS-CoV-2. A number of studies in this regard will be discussed.

In several studies, different structural and functional proteins of coronavirus were targeted using siRNA. Zhang et al. provide evidence that RNAi tool can be a therapeutic approach for SARS-CoV infection. Using spike-specific siRNAs they showed that the siRNAs effectively inhibited the expression of spike protein in cultured cells (85). He et al. evaluated the efficacy of six distinct siRNAs designed for different regions of SARS-CoV replicase gene. Although they did not report any synergistic effects of these siRNA, inhibitory effects of each of the siRNAs were demonstrated in vitro (86). However, later they reported synergistic antiviral activities of siRNAs targeting structural and replicase genes of SARS-associated coronavirus (87). There is also a report suggesting that targeting different regions of SARS-CoV viral genome using siRNA may inhibit viral infection and replication (88). Li et al. demonstrated the potential prophylactic as well as therapeutic ability of SARS coronavirus specific siRNA therapy in Rhesus macaque, with decreased body temperature recorded, lower levels of viral RNA and some decrease in lung damage (89).Overall, there are already several numbers of siRNA related patents regarding the SARS-CoV (90, 91). However, to the best of our knowledge, there are not any active clinical trials using the potential siRNAs for COVID-19.

4.3.2. CRISPR-Cas system

The CRISPR/ Cas system is an adaptive immune system in archaea and bacteria to protect against foreign DNA or in some cases RNA coming from viruses or mobile genetic elements. This immune response consists of three steps: 1. Acquisition of the foreign DNA; during which foreign DNA segments (spacers) are inserted to the genome of the host in between the repeats in CRISPR locus and therefore storing the memory of the previous invader 2. Transcription of the CRISPR locus long transcript and subsequent processing into short CRISPR RNAs (crRNAs) which guides Cas effector proteins to the complementary DNA or RNA sequence in the invading organism 3. Interference happens when the target region is cleaved by a single-protein, Cas effector protein (class 2) or large multisubunit protein complexes (class 1) (92). The CRISPR system comprises two classes and different types and subtypes. Class1 consists of Type I, III and IV and class2 consists of type II (Cas9 effector), V and VI; each having different subtypes. The CRISPR system has been mostly used to perform genome editing and transcription modification. However, this system has been naturally evolved in bacteria to defend against invading phages. This suggests that this system can be repurposed in mammalian cells to defend against RNA and DNA viruses. Type III and Type VI CRISPR systems have RNA targeting activities while the rest target DNA, and therefore they can target RNA and DNA viruses, respectively (93). Successful examples of targeting DNA viruses by CRISPR system type II (Cas9) have been provided in cell culture and animal models for HIV, HBV, herpesvirus, HPV, and many other viruses (94). Recent studies of type IV CRISPR-Cas (effector Cas13) have suggested that they may be able to efficiently target and degrade RNA (95, 96, 97, 98). Therefore, this system provides a potential therapeutic approach for elimination of RNA viruses. Additionally, Cas13 can process the long transcript crRNA and therefore can be used for multiplex targeting. Another advantage of Cas13 is the minimal off target activity on the host transcriptome, as shown in some recent studies (96, 99). Freije et al. showed 94.6% of the 396 single strand RNA (ssRNA) human associated viruses have >10 putative Cas13a target sites (93). Potent Cas13 activity against three different ssRNA viruses (lymphocytic choriomeningitis virus (LCMV); influenza A virus (IAV); and vesicular stomatitis virus (VSV)) was described by Freije et al. The first proof of Cas13 for targeting SARS-CoV-2 has been suggested in the recent study by Abbott et al. In this study, CRISPR-Cas13 was tested as a prophylactic antiviral CRISPR in human cells (PAC-MAN), and showed efficient degradation of SARS-CoV-2 sequences as well as influenza A genome in vitro in human lung epithelial cells (100). This approach could constitute a pan-coronavirus inhibitory strategy, as a group of six cr-RNA might target more than 90% of coronaviruses. Although this study has provided a first proof for the potential usefulness of CRISPR-Cas to target SARS-CoV-2, further studies using replication-incompetent viruses and validation in animal models are required.

5. Immune-based approaches

5.1. Modulating immune responses

5.1.1. Cell based therapy approaches

Mesenchymal stem/stromal cells (MSCs), that can be relatively easily harvested either from fat tissue, bone marrow or placenta, among others, hold the potential for preventing the immune mediated consequences of SARS-CoV-2 and for restoring damaged tissues, by production of both trophic factors and anti-inflammatory molecules (101, 102). These include prostaglandin 2, indoleamine 2,3-dioxygenase (IDO), transforming growth factor-β, HLA-G5, interleukin (IL)-10, nitric oxide, and tumor necrosis factor (TNF)-α-induced gene/ protein 6 (TSG-6) (103). The anti-inflammatory feature of MSCs is exploited for treatment of steroid-resistant, severe, acute graft-versus-host disease (104, 105), and is being tested in the setting of a number of lung inflammatory disorders (106, 107). Moreover, positive effects of SCs in the setting of other viral diseases of the lung have been reported (108). In addition, MSCs have been suggested to potentially ameliorate ARDS, such as what is seen in severely affected COVID-19 patients (109). The immune modulatory role of MSCs in the context of COVID-19 is dual, including antiviral protection mediated by increased interferon stimulated genes (ISGs) expression and a secondary response to IFN, resulting in ISG stimulation and widespread viral resistance (110). Leng et al. showed that infusion of MSCs may improve some laboratory and clinical parameters of patients with COVID-19, and specifically might affect the dysregulated inflammatory responses. For instance, MSC therapy increased peripheral lymphocyte and also regulatory DC cells, while reducing levels of TNF-α and the over-activated cytokine-secreting immune cells, CXCR3+CD4+ T cells, CXCR3+CD8+ T cells and CXCR3+ NK cells (111). Liang et al. reported that the clinical manifestations of a 65- year old woman infected with COVID-19, treated by human umbilical cord MSCs, improved (112).

An alternative approach that might be considered as therapy for COVID-19 related lung disease, are stem cell educator (SCE) therapies. SCE cells are created by culturing patient lymphocytes with cord-blood derived stem cell (CB-SC), after which the autologous lymphocytes can be reinfused into patients. SCE has been demonstrated to potentially restore the balance of immune responses in a variety of autoimmune disease (113). Considering the immunomodulatory properties of SCE cells, researchers are now addressing their efficacy in SARS-related pneumonia (Table 2).

Natural killer (NK) cells are a subset of lymphocytes that exert cytotoxic effects against tumor cells and virus infected cells. In fact, cytokine production (mainly IFNs), by dendritic cells (DCs) stimulates activation of NK cells and these cells perform their cytotoxic activity by secretion of perforin and limiting viral replication (114). Of particular interest is CYNK-001, a NK therapy being developed from placenta derived hematopoietic stem cells (HSC) that is introduced as a potential therapeutic option for a variety of malignancies from hematological to solid tumors (NCT04310592). Celularity Inc., developing allogeneic cellular therapies from human placenta, recently announced the U.S. Food and Drug Administration (FDA) has cleared the company's Investigational New Drug (IND) application for the use of its proprietary CYNK-001 in individuals infected with COVID-19. They will commence a Phase I/II clinical study including up to 86 patients with COVID-19 (Not mentioned in Table 2) (115).

5.2. Anti-cytokine storm therapies

Cytokine release syndrome (CRS), first described in patients received CAR-T cell therapy, characterized by the release of a large body of cytokines including IL-6 and IL-1ß, as well as IL-2, IL-8, IL-17, G- CSF, GM- CSF, IP10, MCP1, MIP1a (also known as CCL3) and TNF (27). TH17 responses, which lead to vascular permeability and leakage, are enhanced by both IL-1β and TNF-α. Moreover, Th-17 shows a wide inflammatory response through producing G-CSF, IL-1β, TNF-α. TH-17 cells are among the cells involved in the COVID-19 cytokine storm. There is evidence that circulating levels of TH-17 cells are increased in COVID-19, suggesting the use of TH-17 blockers as a potential treatment for the present threat, COVID-19. STAT-3 is a transcription factor that mediates inflammatory response of TH-17 cells, while IL-6 activates STAT3 through JAK2 pathway. In the context of COVID-19, Wu et al. proposed that the FDA approved drug, Fedratinib (a JAK2 pathway inhibitor) might be a promising candidate treatment (116). Baricitinb, another member of JAK inhibitors, can improve the clinical status of COVID-19 patients. The related mechanism is by exerting inhibitory effects on numb-associated kinase (NAK) (117).

Type-I interferon (IFN-I) is involved in intracellular pathogen defense in the context of both innate and adaptive immunity. The DNA sensor cyclic GMP–AMP synthase (cGAS) and its downstream effector STING (stimulator of interferon genes) regulate transcription of many inflammatory molecules, including type I and type III interferons. However, dysregulation of IFNs production may lead to inflammatory diseases. Delayed IFN-I production in SARS infected animals, causes the accumulation of pathogenic monocyte-macrophages, resulting in lung immunopathology, vascular leakage, and suboptimal T cell responses. Therefore, targeting cGAS-STING pathway may be a suitable strategy for treatment of severe lung diseases caused by SARS-CoV and SARS-CoV-2. In line with this, Deng et al. evaluated the efficacy of some of the FDA approved drugs for targeting the STING pathway and found that approved drugs, such as suramin and ALK inhibitors, might be efficient and therefore worth testing in clinical trials. Adalimumab (TNF-α) and CMAB806 (IL-6) are among the cytokine-directed antagonists that are in clinical trials for COVID-19 (118). Indeed, anti-IL-6 receptor antibody (anti-IL-6RAb), (chemical name: Tocilizumab), a humanized monoclonal antibody, was developed and showed prophylactic efficacy against a broad spectrum of autoimmune diseases. However, as it was previously mentioned, there is a possibility that as a result of inaccurate attribution of “cytokine storm” with COVID-19, the related therapies would also need reconsideration (45).

Moreover, considering the profound lymphopenia in COVID-19 patients which can lead to severe pathological symptoms, treatments regarding the restoration of lymphocyte numbers have gain attentions. IL-7 has a key role in survival and expansion of lymphocytes. In this regards, Laterre et al. have showed the safety of IL-7 administration in patient with COVID -19 (119).

Interestingly, restoration of Th17/Treg imbalance seems to be the mechanism through which Tocilizumab confers protection over a range of diseases (120, 121). The therapeutic potential of Tocilizumab for treatment of COVID-19 patients is therefore being evaluated. These initial studies suggest that Tocilizumab might revert lymphopenia, lower oxygen need, and improve lung lesion, and therefore suggest that Tocilizumab might be a promising therapeutic strategy for individuals infected with COVID-19 (122). Results of a retrospective cohort study suggest that administered Tocilizumab might mitigate the risk of mechanical ventilation or death in COVID-19 patients with severe pnemonia (123).

5.3. Preventive strategies

5.3.1. Vaccines

Researchers in biotech companies and academic laboratories are making many efforts to develop efficient vaccines against COVID-19 with an unprecedented speed. The number of vaccines that are currently under preclinical investigations confirms this endeavor. A number of approaches have shown sufficiently promising in vitro and in vivo animal results such that they are already moving to early the clinical phase studies (124). In the following, examples of such vaccines discussed. The mRNA-1273 is a lipid nanoparticle encapsulated platform, which encodes the S protein of SARS-CoV-2, and therefore has the potential to elicit a robust immune response (NCT04283461) (125).

Recently, Zhang et al. has developed a novel thermostable mRNA- based vaccine, named ARCoV, which is encoding receptor binding domain of SARS-CoV-2 and elicited a protective response in animals challenged with SARS-CoV-2 (ChiCTR2000034112) (126).

Ad5-nCoV (NCT04313127) and INO-4800 (NCT04336410) vaccine candidates employ the same strategy but using different delivery platforms of adenovirus type 5 and DNA plasmid, respectively. LV-SMENP-DC (NCT04276896) and pathogen-specific aAPC (NCT04299724) are dendritic cells (DCs) and artificial antigen-presenting cells (aAPCs), which are manipulated with lentiviral vector (LV) to express mini-genes based on conserved and critical structural and protease protein domains of virus, respectively. However, developing new vaccination strategies may take months to years in terms of getting approval for both safety and efficacy.

5.3.2. Recombinant nucleic acids

5.3.2.1. DNA vaccines

 

Vaccination is among the most efficient measures against a vast variety of communicable diseases, a permanent global problem. In DNA vaccine technology, the gene encoding an immunogen, is inserted into a suitable vector, which may after administration lead to humoral and / or cellular immunity against this immunogen. Compared with traditional vaccination with an inactivated virus, DNA vaccination holds less risk for infection, is inexpensive, safe and more stable (127). Yang et al. reported that a DNA vaccine encoding for the S glycoprotein of SARS-CoV, exerts T-cell mediated immune response that is accompanied by increased neutralizing antibodies, and protects mice from being infected with SARS-CoV (128). Wang et al. demonstrated that a DNA vaccine encoding for the combination of 2 immunogens, namely the S1 subunit and full-length protein, confers protection against several strains of MERS in non- human primates and mice. Thus, in the context of MERS-CoV, this study is of interest, as the spike protein and S1 subunit DNA vaccine provides potent protection in animal models. Of note, there are some advantages regarding employing spike DNA prime-S1 protein boost over a single protein as the immunogen and for boosting immunization, which involves the production of Th1 immune response along with yielding various neutralizing antibodies (129).

5.3.2.2. mRNA-based strategies

 

An alternative to DNA vaccines, are RNA-based approaches. The problems with instability of RNA, have been in part overcome over the last decade (130). As compared with DNA, RNA-based therapeutics do not suffer from the risk of insertional mutations, and are only short-term present due to the faster RNA degradation. Nowadays, in vitro transcribed (IVT) mRNA has received increasing attention. The mRNA-based drugs are being developed as protein-replacement therapies, immunotherapeutics and in regenerative medicine. Preclinical and clinical applications of IVT-mRNA have been evaluated in a wide range of diseases including viral infections. However, there are some disadvantages regarding these molecules such as their stimulatory effect on immune responses through inducing interferons production. mRNA-1273 is one example of an IVT-mRNA as a novel lipid nanoparticle (LNP)-encapsulated mRNA-based vaccine that encodes for a full-length, spike (S) protein of SARS-CoV-2 (Table 1) (131).

Table 1. An overview of Phase 1, 2 clinical trials regarding gene-based therapy of COVID-19.

 StatusInterventionsPopulationPhaseLocationNCT number
1RecruitingmRNA-1273451•Emory Vaccine Center – The Hope Clinic, Decatur, Georgia, United States
•National Institutes of Health Clinical Center – Vaccine Research Center Clinical Trials Program, Bethesda, Maryland, United States
•Kaiser Permanente Washington Health Research Institute Vaccines and Infectious Diseases, Seattle, Washington, United States
NCT04283461
2RecruitingInjection and infusion of LV-SMENP-DC vaccine and antigen-specific CTLs1001,2•Shenzhen Geno-immune Medical Institute, Shenzhen, Guangdong, China
•Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
•Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
NCT04276896
3RecruitingPathogen specific aAPC1001•Shenzhen Geno-immune Medical Institute, Shenzhen, Guangdong, ChinaNCT04299724
4Active, not recruitingRecombinant Novel Coronavirus Vaccine (Adenovirus Type 5 Vector)1081•Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei, ChinaNCT04313127
5RecruitingINO-4800401•Center for Pharmaceutical Research, Kansas City, Missouri, United States
•University of Pennsylvania, Philadelphia, Pennsylvania, United States
NCT04336410

 

NCT number: The National Clinical Trial number

 

Table 2. An overview of cell-based clinical trials for COVID-19 infections.

1StatusInterventionsPopulationPhaseLocationNCT number
2RecruitingUmbilical cord Wharton's jelly-derived human401,2Hôpital Pitié-Salpêtrière - APHP, Paris, France Hôpital Européen Georges Pompidou - APHP, Paris, FranceNCT04333368
3RecruitingMSC Treatment301,2Istinye University, Istanbul, TurkeyNCT04392778
4RecruitingMSC therapy602,3Royan Institute Tehran, Iran, Islamic Republic ofNCT04366063
5Enrolling byinvitationHB-adMSCs562Hope Biosciences Stem Cell Research Foundation, Sugar Land, Texas, United StatesNCT04349631
6Active, not recruitingUCMSCs1002General Hospital of Central Theater Command, Wuhan, Hubei, China Maternal and Child Hospital of Hubei Province, Wuhan, Hubei, China Wuhan Huoshenshan Hospital, Wuhan, Hubei, ChinaNCT04288102
7RecruitingMSC202Armed Forces Bone Marrow Transplant Centre, Rawalpindi, Punjab, PakistanNCT04444271
8RecruitingHuman cord tissue mesenchymal stromal cells 1,2Duke Hospital, Durham, North Carolina, United StatesNCT04399889
9Not yet recruitingWharton's jelly-MSC401,2Clinical Somer, Rionegro, Antioquia, ColombiaNCT04390152
10Enrolling by invitationAllogenic pooled olfactory mucosa-MSC401,2Institute of Biophysics and Cell Engineering of National Academy of Sciences of Belarus, Minsk, BelarusNCT04382547
11Not yet recruitingMSC402University Hospital Tuebingen, Tuebingen, GermanyNCT04377334
12Not yet recruitingHB-adMSC1002River Oaks Hospital and Clinics, Houston, Texas, United StatesNCT04362189
13RecruitingUmbilical Cord Mesenchymal Stem Cells241,2Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida, United StatesNCT04355728
15Enrolling by invitationHB-adMSCs1002Hope Biosciences Stem Cell Research Foundation, Sugar Land, Texas, United StatesNCT04348435
16Not yet recruitingBMMSCs201,2Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, ChinaNCT04346368
17Not yet recruitingDental pulp mesenchymal stem cells24Early phase 1Not definedNCT04302519
18Not yet recruitingUC-MSCs48Not applicableUnion Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, ChinaNCT04273646
19RecruitingUC-MSCs102Zhongnan Hospital of Wuhan University, Wuhan, Hubei, ChinaNCT04269525
20RecruitingWharton's Jelly-MSCs51Stem Cells Arabia Amman, JordanNCT04313322
21RecruitingMSCs201Beijing 302 Military Hospital of China Beijing, ChinaNCT04252118
22RecruitingUC-MSCs301,2Puren Hospital Affiliated to Wuhan University of Science and Technology Wuhan, Hubei, ChinaNCT04339660
22RecruitingWharton's Jelly-MSCs301,2Hospital del Mar Recruiting Barcelona, Spain, 08003NCT04390139
23RecruitingMSCs3003United StatesNCT04371393
24RecruitingMSCs201,2CHU de Liège Liège, BelgiumNCT04445454
25RecruitingHuman umbilical cord derived CD362 enriched MSCs751,2Belfast Health and Social Care Trust, Royal Hospitals Belfast, Northern Ireland, United KingdomNCT03042143
26RecruitingMSC102Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán Mexico City, MexicoNCT04416139
27RecruitingMSC242Hospital Universitario Rio Hortega Valladolid, SpainNCT04361942
28Not yet recruitingAutologous adipose-derived stem cells2002Armed Forces Bone Marrow Transplant Centre, Rawalpindi, Punjab, PakistanNCT04428801
29RecruitingIntravenous Infusions of Stem Cells202Jinnah Hospital, Lahore, Punjab, PakistanNCT04437823
30Active, not recruitingCOVID-19 Specific T Cell derived exosomes (CSTC Exo)601GENKOK, Kayseri, Melikgazi, TurkeyNCT04389385
31RecruitingCYNK-001861,2Hackensack University Medical Center, Hackensack, New Jersey, United States Atlantic Health, Morristown, New Jersey, United States Atlantic Health, Summit, New Jersey, United States Multicare Health System, Tacoma, Washington, United StatesNCT04365101
32RecruitingNK cell301The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, ChinaNCT04280224
33Not yet recruitingSCE-Treated Mononuclear Cells Apheresis202Not definedNCT04299152
34Not yet recruitingMSCs-derived exosomes301Not definedNCT04276987

 

MSC: Mesenchymal Stem Cell, SCE: Stem Cell Educator

 

5.4. Anticoagulation therapies

Coagulopathy, characterized by high D-dimer levels, increased fibrinogen along with low anti thrombin, is one of the hallmarks associated with patient death due to COVID-19. “Fibrinolytic therapy”, using tissue plasminogen activator (t-PA) could improve survival in animal. models and patients suffering from ARDS. A study by Wand et al. demonstrated that administration of Alteplase (t-PA) was able to improve P/F ratio (the arterial pO2 divided by the fraction of inspired oxygen, expressed as a decimal that the patient is receiving) in COVID-19 patients. However, since the improvement was transient they suggested that re-dosing the anti-fibrotic drug might result in more durable response (132). Disseminated intravascular coagulation (DIC) caused by endothelial dysfunction due to excessive production of thrombin as well as decreased fibrinolysis, is also responsible for COVID-19 lethality. This suggests that anticoagulant agents (e.g. heparin) may be considered as potential anti-COVID-19 candidates. Supporting this idea, Tang et al. showed that anticoagulation therapy using heparin, is presumably associated with a better prognosis in patients infected with COVID-19 (133). In addition, due to some critical features of unfractionated heparin (UFH), also called nebulized heparin, including anti-coagulant, anti-inflammatory and mucolytic effects, UFH may be a potentially effective treatment for COVID-19 (134).

6. Conclusion

The global challenge of the COVID-19 pandemic that is associated with an enormous morbidity and mortality highlights the urgent need for developing efficient therapeutic strategies. Tremendous advances in understanding the molecular basis of the disease pathogenesis in various corona virus-based diseases, and the very fast insights gained in COVID-19 pathogenesis offer opportunities to take a leap in introducing novel and efficient therapeutics as well as preventive measures against COVID-19. Assessing the efficiency of previously approved antiviral agents for inhibiting SARS-CoV-2 is one of the main areas that is being pursued clinically, in the short term. Despite the encouraging results with some of these agents, more research should be done regarding the safety and efficiency, as exemplified by the initial adoption and later withdrawal of the use of chloroquine as a therapy for COVID-19. Different novel therapeutic strategies including cell and gene-based therapies against SARS-CoV-2 are now being developed, of which some are already being tested in early phase clinical trials worldwide. Dysregulation of inflammatory responses, a manifestation of individuals infected with SARS-CoV-2, can be modulated using MSCs. Likewise, SCE cells are another cell based strategy, which will be assessed for its efficiency in relieving SARS- related inflammation. Yet an alternative cell-based strategy is NK cell therapy to inhibit the viral replication. Another approach is based on nucleic acid-based therapies and includes CRISPR methods along with DNA vaccines and mRNA molecules, which are also being intensively studied and some have shown encouraging effects in preclinical trails. Overall, all aforementioned studies inspire further investigation of advanced therapeutic strategies as novel treatment options for COVID-19.

Declaration of Competing Interest

There is nothing to declare

Acknowledgment

The authors would like to express their gratitude to Royan institute and the Katholieke Universiteit Leuven for their support throughout the course of this work.

Funding

The authors received no financial support for authorship, and/or publication of this article.

Ethical Approval

Not required

References

1

L van der Hoek, K Pyrc, MF Jebbink, W Vermeulen-Oost, RJM Berkhout, KC Wolthers, et al.Identification of a new human coronavirus

Nature Medicine, 10 (4) (2004), pp. 368-373

View Record in ScopusGoogle Scholar

2

CI Paules, HD Marston, AS FauciCoronavirus Infections—More Than Just the Common Cold

JAMA, 323 (8) (2020), pp. 707-708

CrossRefView Record in ScopusGoogle Scholar

3

N Zhu, D Zhang, W Wang, X Li, B Yang, J Song, et al.A Novel Coronavirus from Patients with Pneumonia in China, 2019

New England Journal of Medicine., 382 (8) (2020), pp. 727-733

CrossRefView Record in ScopusGoogle Scholar

4

Y Chen, Q Liu, D GuoEmerging coronaviruses: Genome structure, replication, and pathogenesis

Journal of Medical Virology, 92 (4) (2020), pp. 418-423

CrossRefView Record in ScopusGoogle Scholar

5

S Kang, W Peng, Y Zhu, S Lu, M Zhou, W Lin, et al.Recent progress in understanding 2019 novel coronavirus (SARS-CoV-2) associated with human respiratory disease: detection, mechanisms and treatment

Int J Antimicrob Agents (2020), Article 105950

ArticleDownload PDFGoogle Scholar

6

X Ou, Y Liu, X Lei, P Li, D Mi, L Ren, et al.Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV

Nature Communications, 11 (1) (2020), p. 1620

Google Scholar

7

I Sola, F Almazán, S Zúñiga, L EnjuanesContinuous and Discontinuous RNA Synthesis in Coronaviruses

Annu Rev Virol, 2 (1) (2015), pp. 265-288

CrossRefView Record in ScopusGoogle Scholar

8

N Chen, M Zhou, X Dong, J Qu, F Gong, Y Han, et al.Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study

The Lancet., 395 (10223) (2020), pp. 507-513

ArticleDownload PDFView Record in ScopusGoogle Scholar

9

H Xu, L Zhong, J Deng, J Peng, H Dan, X Zeng, et al.High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa

International Journal of Oral Science, 12 (1) (2020), p. 8

CrossRefGoogle Scholar

10

F Hikmet, L Méar, Å Edvinsson, P Micke, M Uhlén, C LindskogThe protein expression profile of ACE2 in human tissues

Molecular Systems Biology, 16 (7) (2020), p. e9610

Google Scholar

11

X-w Pan, D Xu, H Zhang, W Zhou, L-h Wang, X-g. CuiIdentification of a potential mechanism of acute kidney injury during the COVID-19 outbreak: a study based on single-cell transcriptome analysis

Intensive Care Medicine (2020)

Google Scholar

12

F-X Lescure, L Bouadma, D Nguyen, M Parisey, P-H Wicky, S Behillil, et al.Clinical and virological data of the first cases of COVID-19 in Europe: a case series

The Lancet Infectious Diseases (2020)

Google Scholar

13

J Liu, X Zheng, Q Tong, W Li, B Wang, K Sutter, et al.Overlapping and discrete aspects of the pathology and pathogenesis of the emerging human pathogenic coronaviruses SARS-CoV, MERS-CoV, and 2019-nCoV

Journal of Medical Virology, 92 (5) (2020), pp. 491-494

CrossRefView Record in ScopusGoogle Scholar

14

M-L Yeung, Y Yao, L Jia, JFW Chan, K-H Chan, K-F Cheung, et al.MERS coronavirus induces apoptosis in kidney and lung by upregulating Smad7 and FGF2

Nature Microbiology, 1 (3) (2016), p. 16004

Google Scholar

15

H Chu, J Zhou, BH Wong, C Li, JF Chan, ZS Cheng, et al.Middle East Respiratory Syndrome Coronavirus Efficiently Infects Human Primary T Lymphocytes and Activates the Extrinsic and Intrinsic Apoptosis Pathways

The Journal of infectious diseases, 213 (6) (2016), pp. 904-914

CrossRefView Record in ScopusGoogle Scholar

16

R Durvasula, T Wellington, E McNamara, S WatnickCOVID-19 and Kidney Failure in the Acute Care Setting: Our Experience From Seattle

American Journal of Kidney Diseases (2020)

Google Scholar

17

K Alsaad, Y Arabi, A HajeerSpectrum of histopathological findings in coronavirus disease-19, Middle East respiratory syndrome and severe acute respiratory syndrome

Annals of Thoracic Medicine, 15 (2) (2020), pp. 52-53

CrossRefView Record in ScopusGoogle Scholar

18

Z Xu, L Shi, Y Wang, J Zhang, L Huang, C Zhang, et al.Pathological findings of COVID-19 associated with acute respiratory distress syndrome

The Lancet Respiratory Medicine, 8 (4) (2020), pp. 420-422

ArticleDownload PDFView Record in ScopusGoogle Scholar

19

L Wang, Y Zhang, S ZhangCardiovascular Impairment in COVID-19: Learning From Current Options for Cardiovascular Anti-Inflammatory Therapy

Frontiers in Cardiovascular Medicine, 7 (2020), p. 78

CrossRefView Record in ScopusGoogle Scholar

20

S. MusaHepatic and gastrointestinal involvement in coronavirus disease 2019 (COVID-19): What do we know till now?

Arab J Gastroenterol, 21 (1) (2020), pp. 3-8

ArticleDownload PDFView Record in ScopusGoogle Scholar

21

J Zhou, C Li, X Liu, MC Chiu, X Zhao, D Wang, et al.Infection of bat and human intestinal organoids by SARS-CoV-2

Nature Medicine (2020)

Google Scholar

22

B Zhao, C Ni, R Gao, Y Wang, L Yang, J Wei, et al.Recapitulation of SARS-CoV-2 infection and cholangiocyte damage with human liver ductal organoids

Protein Cell (2020), pp. 1-5

CrossRefGoogle Scholar

23

E Prompetchara, C Ketloy, T PalagaImmune responses in COVID-19 and potential vaccines: Lessons learned from SARS and MERS epidemic

Asian Pacific journal of allergy and immunology, 38 (1) (2020), pp. 1-9

View Record in ScopusGoogle Scholar

24

S Khan, R Siddique, MA Shereen, A Ali, J Liu, Q Bai, et al.Emergence of a Novel Coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2: Biology and Therapeutic Options

Journal of Clinical Microbiology, 58 (5) (2020)

e00187-20

Google Scholar

25

R Otsuka, K-i. SeinoMacrophage activation syndrome and COVID-19

Inflammation and Regeneration, 40 (1) (2020), p. 19

Google Scholar

26

M Sokolowska, Z Lukasik, I Agache, CA Akdis, D Akdis, M Akdis, et al.Immunology of COVID-19: mechanisms, clinical outcome, diagnostics and perspectives – a report of the European Academy of Allergy and Clinical Immunology (EAACI)

Allergy (2020)

n/a(n/a)

Google Scholar

27

X. CaoCOVID-19: immunopathology and its implications for therapy

Nature Reviews Immunology (2020)

Google Scholar

28

B Tomar, HJ Anders, J Desai, SR MulayNeutrophils and Neutrophil Extracellular Traps Drive Necroinflammation in COVID-19

Cells, 9 (6) (2020)

Google Scholar

29

X Li, M Geng, Y Peng, L Meng, S LuMolecular immune pathogenesis and diagnosis of COVID-19

Journal of Pharmaceutical Analysis (2020)

Google Scholar

30

S Perlman, AA. DandekarImmunopathogenesis of coronavirus infections: implications for SARS

Nature reviews Immunology, 5 (12) (2005), pp. 917-927

View Record in ScopusGoogle Scholar

31

SF Ahmed, AA Quadeer, MR McKayPreliminary Identification of Potential Vaccine Targets for the COVID-19 Coronavirus (SARS-CoV-2) Based on SARS-CoV Immunological Studies

Viruses, 12 (3) (2020)

Google Scholar

32

A Nguyen, JK David, SK Maden, MA Wood, BR Weeder, A Nellore, et al.Human Leukocyte Antigen Susceptibility Map for Severe Acute Respiratory Syndrome Coronavirus 2

Journal of virology, 94 (13) (2020), pp. e00510-e00520

Google Scholar

33

K Bunte, T. BeiklerTh17 Cells and the IL-23/IL-17 Axis in the Pathogenesis of Periodontitis and Immune-Mediated Inflammatory Diseases

Int J Mol Sci, 20 (14) (2019), p. 3394

CrossRefGoogle Scholar

34

G Li, Y Fan, Y Lai, T Han, Z Li, P Zhou, et al.Coronavirus infections and immune responses

Journal of Medical Virology, 92 (4) (2020), pp. 424-432

CrossRefView Record in ScopusGoogle Scholar

35

OW Ng, A Chia, AT Tan, RS Jadi, HN Leong, A Bertoletti, et al.Memory T cell responses targeting the SARS coronavirus persist up to 11 years post-infection

Vaccine, 34 (17) (2016), pp. 2008-2014

ArticleDownload PDFView Record in ScopusGoogle Scholar

36

G Zhou, Q. ZhaoPerspectives on therapeutic neutralizing antibodies against the Novel Coronavirus SARS-CoV-2

Int J Biol Sci, 16 (10) (2020), pp. 1718-1723

CrossRefView Record in ScopusGoogle Scholar

37

E Traggiai, S Becker, K Subbarao, L Kolesnikova, Y Uematsu, MR Gismondo, et al.An efficient method to make human monoclonal antibodies from memory B cells: potent neutralization of SARS coronavirus

Nat Med, 10 (8) (2004), pp. 871-875

View Record in ScopusGoogle Scholar

38

E Fast, B. ChenPotential T-cell and B-cell Epitopes of 2019-nCoV

bioRxiv., 2020 (2020), Article 955484

02.19

Google Scholar

39

L Liu, Q Wei, Q Lin, J Fang, H Wang, H Kwok, et al.Anti–spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection

JCI Insight, 4 (4) (2019)

Google Scholar

40

Y Imai, K Kuba, S Rao, Y Huan, F Guo, B Guan, et al.Angiotensin-converting enzyme 2 protects from severe acute lung failure

Nature, 436 (7047) (2005), pp. 112-116

CrossRefView Record in ScopusGoogle Scholar

41

C De Miguel, NP Rudemiller, JM Abais, DL MattsonInflammation and hypertension: new understandings and potential therapeutic targets

Curr Hypertens Rep, 17 (1) (2015), p. 507

Google Scholar

42

JC Youn, HT Yu, BJ Lim, MJ Koh, J Lee, DY Chang, et al.Immunosenescent CD8+ T cells and C-X-C chemokine receptor type 3 chemokines are increased in human hypertension

Hypertension (Dallas, Tex: 1979), 62 (1) (2013), pp. 126-133

View Record in ScopusGoogle Scholar

43

Q Ruan, K Yang, W Wang, L Jiang, J SongClinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China

Intensive Care Medicine. (2020)

Google Scholar

44

Q Liu, YH Zhou, ZQ YangThe cytokine storm of severe influenza and development of immunomodulatory therapy

Cell Mol Immunol, 13 (1) (2016), pp. 3-10

CrossRefView Record in ScopusGoogle Scholar

45

P Sinha, MA Matthay, CS CalfeeIs a “Cytokine Storm” Relevant to COVID-19?

JAMA Internal Medicine., 180 (9) (2020), pp. 1152-1154

CrossRefView Record in ScopusGoogle Scholar

46

KE Remy, M Mazer, DA Striker, AH Ellebedy, AH Walton, J Unsinger, et al.Severe immunosuppression and not a cytokine storm characterizes COVID-19 infections

JCI Insight, 5 (17) (2020)

Google Scholar

47

C Sardu, J Gambardella, M Morelli, X Wang, R Marfella, G SantulliIs COVID-19 an endothelial disease?

Clinical and basic evidence. (2020)

Preprints

Google Scholar

48

T Li, H Lu, W ZhangClinical observation and management of COVID-19 patients

Emerging microbes & infections, 9 (1) (2020), pp. 687-690

CrossRefView Record in ScopusGoogle Scholar

49

N Tang, D Li, X Wang, Z SunAbnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia

Journal of Thrombosis and Haemostasis, 18 (4) (2020), pp. 844-847

CrossRefView Record in ScopusGoogle Scholar

50

J-C Song, G Wang, W Zhang, Y Zhang, W-Q Li, Z Zhou, et al.Chinese expert consensus on diagnosis and treatment of coagulation dysfunction in COVID-19

Military Medical Research, 7 (1) (2020), p. 19

View Record in ScopusGoogle Scholar

51

RJ Jose, A. ManuelCOVID-19 cytokine storm: the interplay between inflammation and coagulation

The Lancet Respiratory Medicine (2020)

Google Scholar

52

K Kuba, Y Imai, S Rao, H Gao, F Guo, B Guan, et al.A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury

Nature medicine, 11 (8) (2005), pp. 875-879

CrossRefView Record in ScopusGoogle Scholar

53

F Grillet, J Behr, P Calame, S Aubry, E DelabrousseAcute Pulmonary Embolism Associated with COVID-19 Pneumonia Detected by Pulmonary CT Angiography

Radiology (2020)

201544

Google Scholar

54

L Xiao, H Sakagami, N MiwaACE2: The key Molecule for Understanding the Pathophysiology of Severe and Critical Conditions of COVID-19: Demon or Angel?

Viruses., 12 (5) (2020)

Google Scholar

55

A Heurich, H Hofmann-Winkler, S Gierer, T Liepold, O Jahn, S PöhlmannTMPRSS2 and ADAM17 cleave ACE2 differentially and only proteolysis by TMPRSS2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein

Journal of virology, 88 (2) (2014), pp. 1293-1307

View Record in ScopusGoogle Scholar

56

J Guo, Z Huang, L Lin, J LvCoronavirus Disease 2019 (COVID‐19) and Cardiovascular Disease: A Viewpoint on the Potential Influence of Angiotensin‐Converting Enzyme Inhibitors/Angiotensin Receptor Blockers on Onset and Severity of Severe Acute Respiratory Syndrome Coronavirus 2 Infection

Journal of the American Heart Association, 9 (7) (2020), Article e016219

View Record in ScopusGoogle Scholar

57

K Úri, M Fagyas, A Kertész, A Borbély, C Jenei, O Bene, et al.Circulating ACE2 activity correlates with cardiovascular disease development

Journal of the renin-angiotensin-aldosterone system: JRAAS, 17 (4) (2016)

Google Scholar

58

R Rahmanzade, R Rahmanzadeh, P Tabarsi, SM HashemianNon-Invasive versus Invasive Ventilation in COVID-19: One Size Does Not Fit All!

Anesth Analg. (2020), 10.1213/ANE.0000000000004943

Google Scholar

59

EP Tchesnokov, JY Feng, DP Porter, M GotteMechanism of Inhibition of Ebola Virus RNA-Dependent RNA Polymerase by Remdesivir

Viruses, 11 (4) (2019)

Google Scholar

60

CJ Gordon, EP Tchesnokov, JY Feng, DP Porter, M GotteThe antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus

The Journal of biological chemistry (2020)

Google Scholar

61

ES Amirian, JK. LevyCurrent knowledge about the antivirals remdesivir (GS-5734) and GS-441524 as therapeutic options for coronaviruses

One Health, 9 (2020), Article 100128

ArticleDownload PDFView Record in ScopusGoogle Scholar

62

ML Holshue, C DeBolt, S Lindquist, KH Lofy, J Wiesman, H Bruce, et al.First Case of 2019 Novel Coronavirus in the United States

The New England journal of medicine, 382 (10) (2020), pp. 929-936

CrossRefView Record in ScopusGoogle Scholar

63

JH Beigel, KM Tomashek, LE Dodd, AK Mehta, BS Zingman, AC Kalil, et al.Remdesivir for the Treatment of Covid-19 — Preliminary Report

New England Journal of Medicine (2020)

Google Scholar

64

J Ju, X Li, S Kumar, S Jockusch, M Chien, C Tao, et al.Nucleotide Analogues as Inhibitors of SARS-CoV Polymerase

bioRxiv., 2020 (2020), Article 989186

03.12

Google Scholar

65

L Dong, S Hu, J GaoDiscovering drugs to treat coronavirus disease 2019 (COVID-19)

Drug discoveries & therapeutics, 14 (1) (2020), pp. 58-60

View Record in ScopusGoogle Scholar

66

C Chen, J Huang, P Yin, Y Zhang, Z Cheng, J Wu, et al.Favipiravir versus Arbidol for COVID-19: A Randomized Clinical Trial

medRxiv., 2020 (2020), Article 20037432

03.17

Google Scholar

67

GlenmarkPharmaceuticalsLtd. Glenmark Announces Top-Line Results From Phase 3 Clinical Trial of Favipiravir in Patients With Mild to Moderate COVID-19 2020 [Available from: https://www.prnewswire.com/in/news-releases/glenmark-announces-top-line-results-from-phase-3-clinical-trial-of-favipiravir-in-patients-with-mild-to-moderate-covid-19-886962242.html.

Google Scholar

68

Varadharajan P. Favipiravir Approved as Anti- COVID-19 Drug; DCGI Approves Influenza Treatment Drug For Covid-19 Treatment In India 2020 [Available from: https://www.inventiva.co.in/stories/priyadharshini/favipiravir-approved-as-anti-covid-19-drug-dcgi-approves-influenza-treatment-drug-for-covid-19-treatment-in-india/.

Google Scholar

69

AC Cunningham, HP Goh, D KohTreatment of COVID-19: old tricks for new challenges

Critical Care, 24 (1) (2020), p. 91

CrossRefGoogle Scholar

70

Affairs FsOoM. Coronavirus (COVID-19) Update: FDA Revokes Emergency Use Authorization for Chloroquine and Hydroxychloroquine 2020 [Available from: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-revokes-emergency-use-authorization-chloroquine-and.

Google Scholar

71

JM Molina, C Delaugerre, J Le Goff, B Mela-Lima, D Ponscarme, L Goldwirt, et al.No evidence of rapid antiviral clearance or clinical benefit with the combination of hydroxychloroquine and azithromycin in patients with severe COVID-19 infection

Médecine et Maladies Infectieuses (2020)

Google Scholar

72

AB Cavalcanti, FG Zampieri, RG Rosa, LCP Azevedo, VC Veiga, A Avezum, et al.Hydroxychloroquine with or without Azithromycin in Mild-to-Moderate Covid-19

New England Journal of Medicine (2020)

Google Scholar

73

Chaib F. WHO welcomes preliminary results about dexamethasone use in treating critically ill COVID-19 patients 2020 [Available from: https://www.who.int/news-room/detail/16-06-2020-who-welcomes-preliminary-results-about-dexamethasone-use-in-treating-critically-ill-covid-19-patients.

Google Scholar

74

Dexamethasone in Hospitalized Patients with Covid-19 — Preliminary Report

New England Journal of Medicine. (2020)

Google Scholar

75

AHJ Danser, M Epstein, D BatlleRenin-Angiotensin System Blockers and the COVID-19 Pandemic

Hypertension (Dallas, Tex: 1979) (2020)

0(0):HYPERTENSIONAHA.120.15082

Google Scholar

76

J Clerkin Kevin, A Fried Justin, J Raikhelkar, G Sayer, M Griffin Jan, A Masoumi, et al.Coronavirus Disease 2019 (COVID-19) and Cardiovascular Disease

Circulation, 0 (0) (2020)

Google Scholar

77

M Furuhashi, N Moniwa, T Mita, T Fuseya, S Ishimura, K Ohno, et al.Urinary angiotensin-converting enzyme 2 in hypertensive patients may be increased by olmesartan, an angiotensin II receptor blocker

American journal of hypertension, 28 (1) (2015), pp. 15-21

CrossRefView Record in ScopusGoogle Scholar

78

M Vaduganathan, O Vardeny, T Michel, JJV McMurray, MA Pfeffer, SD SolomonRenin-Angiotensin-Aldosterone System Inhibitors in Patients with Covid-19

The New England journal of medicine, 382 (17) (2020), pp. 1653-1659

CrossRefView Record in ScopusGoogle Scholar

79

D Wrapp, N Wang, KS Corbett, JA Goldsmith, C-L Hsieh, O Abiona, et al.Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation

Science (New York, NY), 367 (6483) (2020), p. 1260

CrossRefView Record in ScopusGoogle Scholar

80

Yuan M, Wu NC, Zhu X, Lee C-CD, So RTY, Lv H, et al. A highly conserved cryptic epitope in the receptor-binding domains of SARS-CoV-2 and SARS-CoV. Science (New York, NY). 2020:eabb7269.

Google Scholar

81

K Duan, B Liu, C Li, H Zhang, T Yu, J Qu, et al.Effectiveness of convalescent plasma therapy in severe COVID-19 patients

Proceedings of the National Academy of Sciences (2020)

202004168

Google Scholar

82

C Shen, Z Wang, F Zhao, Y Yang, J Li, J Yuan, et al.Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma

JAMA (2020)

Google Scholar

83

MJ Joyner, JW Senefeld, SA Klassen, JR Mills, PW Johnson, ES Theel, et al.Effect of Convalescent Plasma on Mortality among Hospitalized Patients with COVID-19: Initial Three-Month Experience

medRxiv., 2020 (2020), Article 20169359

08.12

Google Scholar

84

L Gitlin, S Karelsky, R AndinoShort interfering RNA confers intracellular antiviral immunity in human cells

Nature, 418 (6896) (2002), pp. 430-434

View Record in ScopusGoogle Scholar

85

Y Zhang, T Li, L Fu, C Yu, Y Li, X Xu, et al.Silencing SARS-CoV Spike protein expression in cultured cells by RNA interference

FEBS letters, 560 (1-3) (2004), pp. 141-146

ArticleDownload PDFView Record in ScopusGoogle Scholar

86

ML He, B Zheng, Y Peng, JS Peiris, LL Poon, KY Yuen, et al.Inhibition of SARS-associated coronavirus infection and replication by RNA interference

Jama, 290 (20) (2003), pp. 2665-2666

View Record in ScopusGoogle Scholar

87

ML He, BJ Zheng, Y Chen, KL Wong, JD Huang, MC Lin, et al.Kinetics and synergistic effects of siRNAs targeting structural and replicase genes of SARS-associated coronavirus

FEBS letters, 580 (10) (2006), pp. 2414-2420

ArticleDownload PDFCrossRefView Record in ScopusGoogle Scholar

88

BJ Zheng, Y Guan, Q Tang, C Du, FY Xie, ML He, et al.Prophylactic and therapeutic effects of small interfering RNA targeting SARS-coronavirus

Antiviral therapy, 9 (3) (2004), pp. 365-374

View Record in ScopusGoogle Scholar

89

BJ Li, Q Tang, D Cheng, C Qin, FY Xie, Q Wei, et al.Using siRNA in prophylactic and therapeutic regimens against SARS coronavirus in Rhesus macaque

Nat Med, 11 (9) (2005), pp. 944-951

CrossRefView Record in ScopusGoogle Scholar

90

C Liu, Q Zhou, Y Li, LV Garner, SP Watkins, LJ Carter, et al.Research and Development on Therapeutic Agents and Vaccines for COVID-19 and Related Human Coronavirus Diseases

ACS Cent Sci, 6 (3) (2020), pp. 315-331

CrossRefView Record in ScopusGoogle Scholar

91

S Ghosh, SM Firdous, A NathsiRNA could be a potential therapy for COVID-19

EXCLI J, 19 (2020), pp. 528-531

View Record in ScopusGoogle Scholar

92

G Amitai, R. SorekCRISPR-Cas adaptation: insights into the mechanism of action

Nature reviews Microbiology, 14 (2) (2016), pp. 67-76

CrossRefView Record in ScopusGoogle Scholar

93

CA Freije, C Myhrvold, CK Boehm, AE Lin, NL Welch, A Carter, et al.Programmable Inhibition and Detection of RNA Viruses Using Cas13

Molecular Cell, 76 (5) (2019), pp. 826-837

e11

View Record in ScopusGoogle Scholar

94

C. LeeCRISPR/Cas9-Based Antiviral Strategy: Current Status and the Potential Challenge

Molecules (Basel, Switzerland), 24 (7) (2019)

Google Scholar

95

OO Abudayyeh, JS Gootenberg, S Konermann, J Joung, IM Slaymaker, DB Cox, et al.C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector

Science (New York, NY), 353 (6299) (2016)

aaf5573

Google Scholar

96

OO Abudayyeh, JS Gootenberg, P Essletzbichler, S Han, J Joung, JJ Belanto, et al.RNA targeting with CRISPR–Cas13

Nature, 550 (7675) (2017), pp. 280-284

CrossRefView Record in ScopusGoogle Scholar

97

R Aman, A Mahas, H Butt, F Aljedaani, M MahfouzEngineering RNA Virus Interference via the CRISPR/Cas13 Machinery in Arabidopsis

Viruses, 10 (12) (2018), p. 732

CrossRefGoogle Scholar

98

X Zhao, L Liu, J Lang, K Cheng, Y Wang, X Li, et al.A CRISPR-Cas13a system for efficient and specific therapeutic targeting of mutant KRAS for pancreatic cancer treatment

Cancer letters, 431 (2018), pp. 171-181

ArticleDownload PDFView Record in ScopusGoogle Scholar

99

S Konermann, P Lotfy, NJ Brideau, J Oki, MN Shokhirev, PD HsuTranscriptome Engineering with RNA-Targeting Type VI-D CRISPR Effectors

Cell, 173 (3) (2018), pp. 665-676

View Record in ScopusGoogle Scholar

100

TR Abbott, G Dhamdhere, Y Liu, X Lin, L Goudy, L Zeng, et al.Development of CRISPR as a prophylactic strategy to combat novel coronavirus and influenza

bioRxiv., 2020 (2020), Article 991307

03.13

Google Scholar

101

J Ankrum, JM. KarpMesenchymal stem cell therapy: Two steps forward, one step back

Trends in molecular medicine, 16 (5) (2010), pp. 203-209

ArticleDownload PDFView Record in ScopusGoogle Scholar

102

N Hossein-Khannazer, B Shokoohian, A Shpichka, HA Aghdaei, P Timashev, M VosoughNovel therapeutic approaches for treatment of COVID-19

J Mol Med (Berl), 98 (6) (2020), pp. 789-803

CrossRefView Record in ScopusGoogle Scholar

103

S Elgaz, Z Kuçi, S Kuçi, H Bönig, P BaderClinical Use of Mesenchymal Stromal Cells in the Treatment of Acute Graft-versus-Host Disease

Transfusion Medicine and Hemotherapy, 46 (1) (2019), pp. 27-34

CrossRefView Record in ScopusGoogle Scholar

104

K Le Blanc, F Frassoni, L Ball, F Locatelli, H Roelofs, I Lewis, et al.Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study

Lancet, 371 (9624) (2008), pp. 1579-1586

ArticleDownload PDFView Record in ScopusGoogle Scholar

105

K Muroi, K Miyamura, M Okada, T Yamashita, M Murata, T Ishikawa, et al.Bone marrow-derived mesenchymal stem cells (JR-031) for steroid-refractory grade III or IV acute graft-versus-host disease: a phase II/III study

International journal of hematology, 103 (2) (2016), pp. 243-250

CrossRefView Record in ScopusGoogle Scholar

106

C Emukah, E Dittmar, R Naqvi, J Martinez, A Corral, A Moreira, et al.Mesenchymal stromal cell conditioned media for lung disease: a systematic review and meta-analysis of preclinical studies

Respiratory Research, 20 (1) (2019), p. 239

View Record in ScopusGoogle Scholar

107

J Behnke, S Kremer, T Shahzad, C-M Chao, E Böttcher-Friebertshäuser, R Morty, et al.MSC Based Therapies—New Perspectives for the Injured Lung

Journal of clinical medicine, 9 (2020), p. 682

CrossRefGoogle Scholar

108

J Chen, C Hu, L Chen, L Tang, Y Zhu, X Xu, et al.Clinical study of mesenchymal stem cell treating acute respiratory distress syndrome induced by epidemic Influenza A (H7N9) infection, a hint for COVID-19 treatment

Engineering (Beijing) (2020)

Google Scholar

109

S Horie, HE Gonzalez, JG Laffey, CH MastersonCell therapy in acute respiratory distress syndrome

J Thorac Dis, 10 (9) (2018), pp. 5607-5620

CrossRefView Record in ScopusGoogle Scholar

110

M Khoury, J Cuenca, FF Cruz, FE Figueroa, PRM Rocco, DJ WeissCurrent Status of Cell-Based Therapies for Respiratory Virus Infections: Applicability to COVID-19

European Respiratory Journal (2020)

2000858

Google Scholar

111

Z Leng, R Zhu, W Hou, Y Feng, Y Yang, Q Han, et al.Transplantation of ACE2(-) Mesenchymal Stem Cells Improves the Outcome of Patients with COVID-19 Pneumonia

Aging and disease, 11 (2) (2020), pp. 216-228

CrossRefView Record in ScopusGoogle Scholar

112

Yang, Panrong Nie, Yanfeng Gao, Chuanyun Qian, Min Hu BLJCTLHWWYYLJLCYFNZMMClinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells

ChinaXiv (2020)

Google Scholar

113

Y. ZhaoStem Cell Educator Therapy and Induction of Immune Balance

Current Diabetes Reports, 12 (5) (2012), pp. 517-523

CrossRefView Record in ScopusGoogle Scholar

114

PA Lang, KS Lang, HC Xu, M Grusdat, IA Parish, M Recher, et al.Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity

Proceedings of the National Academy of Sciences, 109 (4) (2012), p. 1210

CrossRefView Record in ScopusGoogle Scholar

115

Celularity I. Celularity Announces FDA Clearance of IND Application for CYNK-001 in Coronavirus, First in Cellular Therapy 2020 [Available from: https://www.prnewswire.com/news-releases/celularity-announces-fda-clearance-of-ind-application-for-cynk-001-in-coronavirus-first-in-cellular-therapy-301034141.html.

Google Scholar

116

D Wu, XO. YangTH17 responses in cytokine storm of COVID-19: An emerging target of JAK2 inhibitor Fedratinib

Journal of Microbiology, Immunology and Infection (2020)

Google Scholar

117

J Stebbing, A Phelan, I Griffin, C Tucker, O Oechsle, D Smith, et al.COVID-19: combining antiviral and anti-inflammatory treatments

The Lancet Infectious Diseases, 20 (4) (2020), pp. 400-402

ArticleDownload PDFView Record in ScopusGoogle Scholar

118

Xiaobing D, Yu X, Pei J. Regulation of interferon production as a potential strategy for COVID-19 treatment2020.

Google Scholar

119

PF Laterre, B François, C Collienne, P Hantson, R Jeannet, KE Remy, et al.Association of Interleukin 7 Immunotherapy With Lymphocyte Counts Among Patients With Severe Coronavirus Disease 2019 (COVID-19)

JAMA Network Open, 3 (7) (2020)

e2016485-e

Google Scholar

120

T Tanaka, M Narazaki, T KishimotoTherapeutic targeting of the interleukin-6 receptor

Annual review of pharmacology and toxicology, 52 (2012), pp. 199-219

CrossRefView Record in ScopusGoogle Scholar

121

S Kang, T Tanaka, T KishimotoTherapeutic uses of anti-interleukin-6 receptor antibody

International immunology, 27 (1) (2015), pp. 21-29

CrossRefView Record in ScopusGoogle Scholar

122

MHTL Xiaoling Xu, Wei Sun, Dongsheng Wang, Binqing Fu, Yonggang Zhou, Xiaohu Zheng, Yun Yang, Xiuyong Li, Xiaohua Zhang, Aijun Pan, Haiming WeiEffective Treatment of Severe COVID-19 Patients with Tocilizumab

chinaXiv. (2020), pp. 1-12

Google Scholar

123

G Guaraldi, M Meschiari, A Cozzi-Lepri, J Milic, R Tonelli, M Menozzi, et al.Tocilizumab in patients with severe COVID-19: a retrospective cohort study

The Lancet Rheumatology, 2 (8) (2020), pp. e474-ee84

View Record in ScopusGoogle Scholar

124

ZA Tung Thanh Le, Arun Kumar, Raúl Gómez Román, Stig Tollefsen, Melanie Saville, Stephen MayhewThe COVID-19 vaccine development landscape

Nature Reviews Drug Discovery (2020)

Google Scholar

125

LA Jackson, EJ Anderson, NG Rouphael, PC Roberts, M Makhene, RN Coler, et al.An mRNA Vaccine against SARS-CoV-2 - Preliminary Report

The New England journal of medicine (2020)

Google Scholar

126

N-N Zhang, X-F Li, Y-Q Deng, H Zhao, Y-J Huang, G Yang, et al.A Thermostable mRNA Vaccine against COVID-19

Cell, 182 (5) (2020), pp. 1271-1283

e16

CrossRefView Record in ScopusGoogle Scholar

127

KH KhanDNA vaccines: roles against diseases

Germs, 3 (1) (2013), pp. 26-35

CrossRefView Record in ScopusGoogle Scholar

128

ZY Yang, WP Kong, Y Huang, A Roberts, BR Murphy, K Subbarao, et al.A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice

Nature, 428 (6982) (2004), pp. 561-564

View Record in ScopusGoogle Scholar

129

L Wang, W Shi, MG Joyce, K Modjarrad, Y Zhang, K Leung, et al.Evaluation of candidate vaccine approaches for MERS-CoV

Nature Communications, 6 (1) (2015), p. 7712

Google Scholar

130

P Guo, F Haque, B Hallahan, R Reif, H LiUniqueness, advantages, challenges, solutions, and perspectives in therapeutics applying RNA nanotechnology

Nucleic Acid Ther, 22 (4) (2012), pp. 226-245

View Record in ScopusGoogle Scholar

131

U Sahin, K Karikó, Ö. TürecimRNA-based therapeutics — developing a new class of drugs

Nature Reviews Drug Discovery, 13 (10) (2014), pp. 759-780

CrossRefView Record in ScopusGoogle Scholar

132

J Wang, N Hajizadeh, EE Moore, RC McIntyre, PK Moore, LA Veress, et al.Tissue Plasminogen Activator (tPA) Treatment for COVID-19 Associated Acute Respiratory Distress Syndrome (ARDS): A Case Series

Journal of thrombosis and haemostasis: JTH (2020)

Google Scholar

133

N Tang, H Bai, X Chen, J Gong, D Li, Z SunAnticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy

Journal of Thrombosis and Haemostasis, 18 (5) (2020), pp. 1094-1099

CrossRefView Record in ScopusGoogle Scholar

134

FMP van Haren, C Page, JG Laffey, A Artigas, M Camprubi-Rimblas, Q Nunes, et al.Nebulised heparin as a treatment for COVID-19: scientific rationale and a call for randomised evidence

Critical Care, 24 (1) (2020), p. 454

Google Scholar

1

Roya Ramezankhani and Roya Solhi contributed equally to this work.

View Abstract

© 2020 Elsevier Ltd and International Society of Antimicrobial Chemotherapy. All rights reserved.