Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1498 2022-11-02 03:54:48 |
2 format corrected. Meta information modification 1498 2022-11-02 07:25:25 | |
3 format corrected. + 2 word(s) 1500 2022-11-02 07:26:41 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ho, C.;  Lee, P. Advances in COVID-19 Treatment. Encyclopedia. Available online: https://encyclopedia.pub/entry/32384 (accessed on 19 April 2024).
Ho C,  Lee P. Advances in COVID-19 Treatment. Encyclopedia. Available at: https://encyclopedia.pub/entry/32384. Accessed April 19, 2024.
Ho, Chian, Ping-Chin Lee. "Advances in COVID-19 Treatment" Encyclopedia, https://encyclopedia.pub/entry/32384 (accessed April 19, 2024).
Ho, C., & Lee, P. (2022, November 02). Advances in COVID-19 Treatment. In Encyclopedia. https://encyclopedia.pub/entry/32384
Ho, Chian and Ping-Chin Lee. "Advances in COVID-19 Treatment." Encyclopedia. Web. 02 November, 2022.
Advances in COVID-19 Treatment
Edit

COVID-19, which emerged in December 2019, was declared a global pandemic by the World Health Organization (WHO) in March 2020. The disease was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has caused millions of deaths worldwide and caused social and economic disruption. While clinical trials on therapeutic drugs are going on in an Accelerating COVID-19 Therapeutic Interventions and Vaccines (ACTIV) public–private partnership collaboration, current therapeutic approaches and options to counter COVID-19 remain few. Therapeutic drugs include the FDA-approved antiviral drugs, Remdesivir, and an immune modulator, Baricitinib. Hence, therapeutic approaches and alternatives for COVID-19 treatment need to be broadened. 

COVID-19 peptides virus life cycle FDA-approved drugs drug development

1. Expanding the Therapeutic Approaches for COVID-19

Despite significant efforts to produce a successful COVID-19 treatment, there are now only a small number of FDA-approved therapeutic medicines for COVID-19 therapy. Remdesivir and baricitinib are the only available treatment medications. It would be advantageous to investigate additional possibilities, such as small compounds and peptides, for developing COVID-19 medicines. The therapeutic approaches also focus on boosting the host’s immune system with monoclonal antibodies, inhibiting virus genome replication by inhibiting RdRp, and prescribing anti-inflammatory drugs and immune modulators to reduce the host’s hyper inflammation response. Other strategies, such as preventing viral entry into host cells or inhibiting the formation of RTC through inhibiting the main protease, should also be considered.

1.1. Peptides Targeting S Protein RBD

Several strategies prevent viral entry into host cells, including blocking  receptor-binding domain (RBD) and ACE2 binding, preventing S protein cleavage, and blocking the binding of HR1 and HR2 by small compounds and peptides. In 2020, Cao et al. designed several mini proteins, AHB1, AHB2, LCB1, and LCB3, which bind to the RBD of the SARS-CoV-2 S protein. These proteins inhibit infection of SARS-CoV-2 to Vero E6 cells with IC50 of 35 nM, 15.5 nM, 23.5 pM, and 48.1 pM, respectively [1]. However, these mini proteins are a few folds larger than standard peptides, and there is no information on in vivo models. Wolfe et al. generated a list of peptides using computer modeling and further tested their binding affinity using bio-layer interferometry (BLI). They highlighted four peptides, P89, P100, P168, and P180, which bind to subunit 1 of S protein with binding constants (KD) 124 nM, 185 nM, 143 nM, and 243 nM, respectively [2]. However, there is no information on additional analysis, such as IC50 determination or in vivo study.
The most promising stapled peptide created by Curelli et al. to target the S protein RBD was NYBSP-4. NYBSP-4 has a KD value of 2.2 µM using surface plasmon resonance (SPR). It expressed a calculated half-life (T1/2) of >289 min in human plasma, showing high proteolytic stability. It did not demonstrate toxicity even when tested with a maximum dose and has an IC50 of 1.97 µM and 2.8 µM in two cell types, HT1080/ACE2 and A549/ACE2 cells, upon infection with the SARS-CoV-2 pseudovirus [3]. However, there is no investigation using in vivo models. In addition, Karoyan et al. designed three interesting peptides, P8, P9, and P10, with IC50 values of 46 nM, 53 nM, and 42 nM on Calu-3 cells following SARS-CoV-2 infection with KD values of 24 nM, 0.09 nM, and 0.03 nM, respectively. These peptides did not cause any cytotoxicity in Vero-E5 and Calu-3 cells [4]. However, there is no testing of these peptides in vivo. Among the list of peptides developed by Chen et al. in 2021 is a peptide called AYn1, which has a KD value of 95.6 nM using localised surface plasmon resonance (LSPR) and an IC50 value of 4.9 μM when infecting HEK293T/hACE2 with SARS-CoV-2 pseudovirus and demonstrated minimal cytotoxicity to the cells. However, the result of AYn1 in in vivo models is not satisfactory [5].

1.2. Peptides Targeting S Protein HR1-HR2 Fusion

Several peptides have been reported to inhibit the fusion of HR1 and HR2 of SARS-CoV-2 [6]. One peptide, EK1C4, can inhibit infection of SARS-CoV-2 pseudotyped and live virus at IC50 of 15.8 nM and 36.5 nM, respectively. EK1C4 is found to exhibit little or no toxic effect in vitro. The modified EK1C4 peptide, EKL1C, has an inhibitory effect on SARS-CoV-2 pseudotyped and live virus infection at IC50 of 3–27 nM in various cell types. EKL1C also demonstrated the ability to reduce SARS-CoV-2 virus titer in the lungs of hACE2-Tg mice. It has stronger resistance to proteolytic enzymes and exhibits higher thermostability properties. A dimeric peptide candidate, [SARSHRC-PEG4]2-chol lipopeptide, has been reported to inhibit HR1 and HR2 fusion. It inhibits viral entry after 8 h on Vero-E6 and Vero-E6-TMPRSS2 cells with IC50 of ~300 nM and ~5 nM, respectively, and showed no toxicity in a cellular toxicity assay [7].

1.3. Small Molecules and Peptides Targeting Host Proteases and SARS-CoV-2 Main Protease

Several small molecules and peptides that target host proteases have been reported. The TMPRSS2 peptide mimetic inhibitors MI-432, MI-1900, the peptide aprotinin, and the furin inhibitor MI-1851 all suppress SARS-CoV-2 replication in Calu-3 [8]. Apart from this, furin/proprotein convertase (PC) inhibitors, decanoyl-RVKR-chloromethylketone (CMK), naphthofluorescein, and a TMPRSS2 inhibitor, camostat effectively decreases SARS-CoV-2 production in Vero-E6 cells [9]. Teicoplanin, a glycopeptide antibiotic that inhibits cathepsin L, effectively halted the infection of HEK293T and Huh7 cells by SARS-CoV-2 pseudotyped viruses [10]. However, further analyses such as cytotoxicity or in vivo model tests have not been carried out in these studies.
An in vitro fluorescence resonance energy transfer (FRET) assay identified five drugs that are inhibitors of the SARS-CoV-2 main protease and have IC50 values of 4.81 µM, 5.4 µM, 16.2 µM, 38.5 µM, and 18.7 µM, respectively. These drugs are manidipine, boceprevir, lercanidipine, efonidipine, and bedaquiline [11]. In another study, four peptides, p12, p13, p15, and p16, showed binding to the main protease and competitively inhibiting main protease activity. The peptides have an IC50 value of 5.36 µM, 3.11 µM, 5.31 µM, and 3.76 µM, respectively, as determined via solid-phase extraction coupled to mass spectrometry (SPE MS) [12]. In Vero cells infected with SARS-CoV-2, two other drugs, ebselen and a Michael acceptor inhibitor known as N3, showed antiviral effects [13]. This suggests that these two drugs may be able to permeate the host cell membrane and target the primary protease. Teicoplanin, also has inhibitory activity on the SARS-CoV-2 main protease [14]. However, further analyses, such as cytotoxicity or in vivo models, are also lacking.

2. A New Area That Can Be Considered for the Development of COVID-19 Drugs

There is an area that remained seemingly untouched in the therapeutic approach that prevents viral entry into host cells. Apart from inhibiting host cell proteases and the primary SARS-CoV-2 protease and preventing the interaction of the RBD with ACE2 and the HR1-HR2 fusion, studies on developing therapeutic drugs that can bind to the S protein’s S1/S2 cleavage site and stop host proteases from breaking it down should be considered. The S protein has an N-terminal domain (residue 14–305), RBD (residue 319–541), S1/S2 cleavage site where host proteases cleave between residue 685 and 686, fusion peptide domain (residue 788–806), HR1 (residue 912–984), HR2 (residue 1163–1213), transmembrane domain (residue 1213–1237), and cytoplasm domain (residue 1237–1273) [15]. Compared to the RBD, the S protein’s S1/S2 cleavage site is relatively conserved, as shown in Table 1 [16]. For this reason, therapeutic drugs that target S1/S2 cleavage site might face fewer challenges caused by mutation of the targeted binding site.
Developing therapeutic drugs targeting the S1/S2 cleavage site instead of host proteases seems more applicable, as the host proteases are involved in important physiological processes. Consider the PC family member furin, which is a key player in embryogenesis. Experimenting on furin knockout mice results in embryonic fatality. Furin also aids in synaptic innervation, which is another crucial role. It is accomplished by the pro-nerve growth factor’s pro-NGF neurotrophin, created by furin cleaving it and binding to the Trk receptors (NGF). Pro-NGF is secreted when furin is inhibited, and it binds to the neurotrophin receptor (p75NTR) and triggers apoptosis [17]. In addition, Cathepsin L is involved in proteolytic activities and generates active enzymes, receptors, and biologically active peptides [18].
Furthermore, furin belongs to the PC family, TMPRSS2, the transmembrane-bound serine proteases family, and cathepsin L, the cathepsin family. Proteases within a common family possess similar structures and mechanisms. Hence, the inhibitors targeting these proteases might also end up targeting proteases of the same family, disrupting important physiological functions with unforeseen consequences. As a result, targeting the S protein’s S1/S2 cleavage site might be a better course of action than suppressing the host proteases.

References

  1. Cao, L.; Goreshnik, I.; Coventry, B.; Case, J.B.; Miller, L.; Walls, A.C.; Park, Y.; Strauch, E.; Stewart, L.; Diamond, M.S.; et al. De novo design of picomolar SARS-CoV-2 miniprotein inhibitors. Science 2020, 370, 426–431.
  2. Wolfe, M.; Webb, S.; Chushak, Y.; Krabacher, R.; Liu, Y.; Swami, N.; Harbaugh, S.; Chávez, J. A high-throughput pipeline for design and selection of peptides targeting the SARS-CoV-2 Spike protein. Sci. Rep. 2021, 11, 21768.
  3. Curreli, F.; Victor, S.M.B.; Ahmed, S.; Drelich, A.; Tong, X.; Tseng, C.T.K.; Hillyer, C.; Debnath, A.K. Stapled peptides based on human angiotensin-converting enzyme 2 (ACE2) potently inhibit SARS-CoV-2 infection in vitro. MBio 2020, 11, e02451-20.
  4. Karoyan, P.; Vieillard, V.; Gómez-Morales, L.; Odile, E.; Guihot, A.; Luyt, C.E.; Denis, A.; Grondin, P.; Lequin, O. Human ACE2 peptide-mimics block SARS-CoV-2 pulmonary cells infection. Commun. Biol. 2021, 4, 197.
  5. Chen, J.; Li, S.; Lei, Z.; Tang, Q.; Mo, L.; Zhao, X.; Xie, F.; Zi, D.; Tan, J. Inhibition of SARS-CoV-2 pseudovirus invasion by ace2 protecting and spike neutralizing peptides: An alternative approach to covid19 prevention and therapy. Int. J. Biol. Sci. 2021, 17, 2957–2969.
  6. Xia, S.; Liu, M.; Wang, C.; Xu, W.; Lan, Q.; Feng, S.; Qi, F.; Bao, L.; Du, L.; Liu, S.; et al. Inhibition of SARS-CoV-2 (previously 2019-nCoV) infection by a highly potent pan-coronavirus fusion inhibitor targeting its spike protein that harbors a high capacity to mediate membrane fusion. Cell Res. 2020, 30, 343–355.
  7. Vries, R.D.; De Schmitz, K.S.; Bovier, F.T.; Predella, C.; Khao, J.; Noack, D.; Haagmans, B.L.; Herfst, S.; Stearns, K.N.; Drew-Bear, J.; et al. Intranasal fusion inhibitory lipopeptide prevents direct-contact SARS-CoV-2 transmission in Ferrets. Science 2021, 371, 1379–1382.
  8. Bestle, D.; Heindl, M.R.; Limburg, H.; van Lam, T.V.; Pilgram, O.; Moulton, H.; Stein, D.A.; Hardes, K.; Eickmann, M.; Dolnik, O.; et al. TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells. Life Sci. Alliance 2020, 3, e202000786.
  9. Cheng, Y.W.; Chao, T.L.; Li, C.L.; Chiu, M.F.; Kao, H.C.; Wang, S.H.; Pang, Y.H.; Lin, C.H.; Tsai, Y.M.; Lee, W.H.; et al. Furin Inhibitors Block SARS-CoV-2 Spike Protein Cleavage to Suppress Virus Production and Cytopathic Effects. Cell Rep. 2020, 33, 108254.
  10. Zhang, J.; Ma, X.; Yu, F.; Liu, J.; Zou, F.; Pan, T.; Zhang, H. Teicoplanin potently blocks the cell entry of 2019-nCoV. bioRxiv 2020.
  11. Ghahremanpour, M.M.; Tirado-Rives, J.; Deshmukh, M.; Ippolito, J.A.; Zhang, C.H.; Cabeza De Vaca, I.; Liosi, M.; Anderson, K.S.; Jorgensen, W.L. Identification of 14 Known Drugs as Inhibitors of the Main Protease of SARS-CoV-2. ACS Med. Chem. Lett. 2020, 11, 2526–2533.
  12. Chan, H.T.H.; Moesser, M.A.; Walters, R.K.; Malla, T.R.; Twidale, R.M.; John, T.; Deeks, H.M.; Johnston-Wood, T.; Mikhailov, V.; Sessions, R.B.; et al. Discovery of SARS-CoV-2 Mpropeptide inhibitors from modelling substrate and ligand binding. Chem. Sci. 2021, 12, 13686–13703.
  13. Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; et al. Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289–293.
  14. Tripathi, K.P.; Upadhyay, S.; Singh, M.; Raghavendhar, S.; Bhardwaj, M. Screening and evaluation of approved drugs as inhibitors of main protease of SARS-CoV-2. Int. J. Biol. Macromol. 2020, 164, 2622–2631.
  15. Huang, Y.; Yang, C.; Xu, X.F.; Xu, W.; Liu, S.W. Structural and functional properties of SARS-CoV-2 spike protein: Potential antivirus drug development for COVID-19. Acta Pharmacol. Sin. 2020, 41, 1141–1149.
  16. SARS-CoV-2 Variants of Concern as of 25 May 2022. Available online: https://www.ecdc.europa.eu/en/covid-19/variants-concern (accessed on 31 May 2022).
  17. Thomas, G. Furin at the cutting edge: From protein traffic to embryogenesis and disease. Mol. Cell 2007, 3, 753–766.
  18. Kirschke, H.; Cathepsin, L. Handbook of Proteolytic Enzymes, 3rd ed.; Rawlings, N.D., Salvesen, G., Eds.; Academic Press: London, UK, 2013; Volume 2, pp. 1808–1817.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 379
Entry Collection: COVID-19
Revisions: 3 times (View History)
Update Date: 02 Nov 2022
1000/1000