Open access

Introductory Chapter: Current Perspective of COVID-19 Drugs

Written By

Arli Aditya Parikesit and Rizky Nurdiansyah

Published: 07 December 2022

DOI: 10.5772/intechopen.105537

From the Edited Volume

COVID-19 Drug Development - Recent Advances, New Perspectives and Applications

Edited by Arli Aditya Parikesit

Chapter metrics overview

67 Chapter Downloads

View Full Metrics

1. Introduction

1.1 Early efforts of drug repurposing

When WHO declared that the COVID-19 pandemic was in place on March 2020, there were no standard drugs available for this new disease. One of the fastest and the most effective solution to face this problem is relying on the drug repurposing effort. Remdesivir is one of the earliest repurposed drug, as it has been originally develop for hepatitis C, and already examined for application in Ebola and Marburg virus infection [1, 2]. In the laboratory setting, remdesivir has been proven to inhibit the RdRp protein of SARS-CoV-2 that is responsible for viral replication, shortens hospitalization period, and is supported with vivid computational model [2, 3, 4, 5]. Hence, remdesivir could only be provided in hospital as it should be delivered intravenously to the patients, thus deterring out-patient application [6]. In parallel, some other repurposing efforts are on the way as well.

These anti-worm and anti-parasitic drugs, Ivermectin, Chloroquine, and Hydroxychloroquine, are also repurposed. Although they provide promising in vitro and in vivo leads, there are inconclusive clinical trial results. Moreover, some meta-analysis results provide some leads that are yet to be validated in the clinical trials [7, 8, 9, 10, 11, 12]. Hence, there are some repurposed drugs that are efficacious to reduce the severe effects of COVID-19, although they are not anti-viral ones. Dexamethasone, a long-standing standard drug of anti-inflammation and anti-coagulation, could inhibit the ‘cytokine storm’ of late stage COVID-19 [13]. It also acts as immune modulator as well [14, 15]. Then, Heparin that works as anti-blot clot drug, also deployed to the COVID-19 patients [16, 17, 18, 19]. However, during the early phase of COVID-19 pandemics, there are no standard drugs that could provide solid clinical evidence and could be deployed in out-patients.

Advertisement

2. Ascendancy of standard drugs for COVID-19

The development of Paxlovid and Molnupiravir, both of them are synthetic drugs, have enabled new standard due to the emergency FDA’s authorization [20, 21]. As nucleoside analogue, molnupiravir has successfully inhibited RdRp protein of SARS-CoV-2 while providing significant clinical trials result for patients with mild-to-moderate symptoms of COVID-19 [22]. Thus, Paxlovid, which is a combination of two distinctive drugs of nirmatrelvir dan ritonavir, could inhibit SARS-CoV-2’s main protease enzyme with retaining its acceptable concentration in the blood, and reduces hospital admission alongside with mortality rate significantly [23, 24]. Hence, both drugs were optimized with rational drug design methods and approved by the FDA with emergency authorization [22, 23, 24, 25, 26]. Although remdesivir has provided significant efficacy in the clinical setting, its in-patient setting prevented the wide-spread use. However, the development of oral remdesivir has been devised and the current status is deemed successful in the in vivo trials [27, 28]. Although careful and thorough examination by medical specialist is necessary before deploying those anti-viral drugs, they are considered the available standard drugs for COVID-19 so far.

Advertisement

3. FAERS database annotation of COVID-19 drug trials

FDA Adverse Event Reporting System (FAERS) Public Dashboard is the Bioinformatics approach in curating side effects of drug trials from clinical data [29, 30]. It has special public dashboard for COVID-19 emergency use authorization (EUA) products, as seen in the Figure 1.

Figure 1.

COVID-19 emergency use authorization (EUA) FAERS public dashboard (source: https://www.fda.gov/drugs/questions-and-answers-fdas-adverse-event-reporting-system-faers/fda-adverse-event-reporting-system-faers-public-dashboard).

The FAERS database has annotated COVID-19 drug trials side effects, such as the trials with hydroxychloroquine/chloroquine, bamlanivimab, Ribavirin-Interferon and ivermectin [31, 32, 33, 34, 35, 36, 37]. Moreover, cancer patients who were old (65 year-old and above), males, and taking immunosuppressive treatment, as well as those with hematological malignancies, were at a higher risk of death due to COVID-19 infection, according to FAERS data [34]. Hence, pertaining the existing COVID-19 standard drugs, the finding in FAERS database did not affect current FDA’s endorsement as it is considered still a relatively novel development. More trials will be necessary. Thus, extensive computational and wet lab studies of the leads’ pharmacological and toxicological properties should be carefully examined [38].

Advertisement

4. Promise of natural product leads

Since the early phase of the pandemics, many researchers have tried to examine bioactive compounds such as propolis and others, from natural products source as leads for SARS-CoV-2 drug candidates [39, 40, 41, 42, 43, 44, 45, 46, 47]. Although these efforts are still on going up to now, the leads are still in experimental stage in general. Several leads were developed with computational approaches from natural products, albeit still no conclusive experimental evidence that leads into clinical trials [48, 49, 50, 51]. There is also effort for repurposing natural product leads, from H5N1 inhibitor lead compounds, that are yet to provide wet lab results [52]. One example of the lead compound with in silico potential for inhibiting SARS-CoV-2 is Juglanin, as visualized in Figure 2.

Figure 2.

Juglanin, provided as the lead for SARS-CoV-2 drug candidate based on in silico approach [53] (source: https://pubchem.ncbi.nlm.nih.gov/compound/5318717#section=2D-Structure).

Computation of natural products chemistry is a way to leverage the indigenous knowledge with modern science such as bioinformatics and biomedicine [48, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63]. It has achieve certain success in medical application, such as the chemotherapy agent of Taxol [64, 65]. However, there is no standard natural product-based drug yet for COVID-19 up to now. In general, existing COVID-19 standard drugs are repurposed drugs that mainly comprises of synthetic and semi-synthetic compounds [66, 67]. How natural products bioactive compounds will perform in this domain, it remains to be seen.

Advertisement

5. Coronavirus and anticipation of future pandemics

The SARS, MERS, and COVID-19 pandemics have taught us valuable lessons that coronavirus outbreak could not be underestimated. This particular virus has showcased themselves as constant menace to the public health. Therefore, it would need establishment of a dedicated research network world-wide to cater this challenge. The current pandemics has been predicted in quite a while. Based on WHO guidelines in 2015, it is stated explicitly that coronavirus has a prospect of becoming pandemic [68, 69]. There are hundreds or even thousands coronavirus species that potentially some of them could be spilled to human population from animals [68, 69]. Although conclusive evidence is still gathered on the moment, the existing leads are pointing to the spill over of coronavirus from bat as precursor of this on-going pandemic [70, 71, 72, 73]. As a RNA virus, it has high mutation rate for the genetic material, and as new SARS-CoV-2 variants emerged, it elicits potential to tamper the efficacy of the existing therapy [74, 75, 76, 77]. In these uncertain conditions, Biologics and/or biosimilars such as monoclonal antibody, polypeptide, and protein-based therapy are proposed as potential therapies for COVID-19 infections. Their high bioavailability in the cell is one of the strength in the pharmacokinetics properties [78]. Monoclonal antibodies therapy has been proven to provide good efficacy against COVID-19 in clinical setting [79, 80]. Moreover, some peptide-based leads are currently under development as COVID-19 drug candidates [81]. For siRNA, some in silico and wet lab research are underway, albeit the clinical application is yet to be determined [82, 83, 84]. In this end, more variative approaches are available as means to anticipate future coronavirus pandemics.

Advertisement

Acknowledgments

The authors would like to express their gratitude to the Indonesia International Institute for Life Sciences (i3L) Department of Research and Community Service (LPPM) for their support of this study. The authors wish to express their gratitude to the Direktorat Sumber Daya, Kementerian Pendidikan, Kebudayaan, dan Ristek, as well as LLDIKTI3, for giving Hibah Penelitian Dasar Berbasis Kompetensi 2022 No. 0054/E5/AK.04/2022.

References

  1. 1. Malin JJ, Suárez I, Priesner V, Fätkenheuer G, Rybniker J. Remdesivir against COVID-19 and other viral diseases. Clinical Microbiology Reviews. 2021;34(1):1-21. DOI: 10.1128/CMR.00162-20
  2. 2. Eastman RT et al. Remdesivir: A review of its discovery and development leading to emergency use authorization for treatment of COVID-19. ACS Central Science. 2020;6(5):672. DOI: 10.1021/ACSCENTSCI.0C00489
  3. 3. Parikesit AA, Nurdiansyah R. Drug repurposing option for COVID-19 with structural bioinformatics of chemical interactions approach. Cermin Dunia Kedokteran. 2020;47(3):222-226. DOI: 10.5281/zenodo.4460736
  4. 4. Shannon A et al. Remdesivir and SARS-CoV-2: Structural requirements at both nsp12 RdRp and nsp14 exonuclease active-sites. Antiviral Research. 2020;178:104793. DOI: 10.1016/j.antiviral.2020.104793
  5. 5. Arba M, Wahyudi ST, Brunt DJ, Paradis N, Wu C. Mechanistic insight on the remdesivir binding to RNA-dependent RNA polymerase (RdRp) of SARS-cov-2. Computers in Biology and Medicine. 2021;129:104156. DOI: 10.1016/j.compbiomed.2020.104156
  6. 6. Gottlieb RL et al. Early Remdesivir to prevent progression to severe Covid-19 in outpatients. New England Journal of Medicine. 2021;386(4):305-315. DOI: 10.1056/NEJMOA2116846
  7. 7. Chosidow O et al. Ivermectin as a potential treatment for COVID-19? PLoS Neglected Tropical Diseases. 2021;15(6):e0009446. DOI: 10.1371/journal.pntd.0009446
  8. 8. Food and Drug Administration (FDA). Why You Should Not Use Ivermectin to Treat or Prevent COVID-19. Coronavirus. 2021. Available from: https://www.fda.gov/consumers/consumer-updates/why-you-should-not-use-ivermectin-treat-or-prevent-covid-19 [Accessed: June 10, 2021]
  9. 9. WHO. WHO Advises that Ivermectin only be used to Treat COVID-19 Within Clinical Trials. Feature stories. 2021. Available from: https://www.who.int/news-room/feature-stories/detail/who-advises-that-ivermectin-only-be-used-to-treat-covid-19-within-clinical-trials [Accessed: June 10, 2021]
  10. 10. Meyerowitz-Katz G, Wieten S, Arellano M d JM, Yamey G. Unethical studies of ivermectin for covid-19. BMJ. 2022;377:o917. DOI: 10.1136/bmj.o917
  11. 11. Ahmed S et al. A five-day course of ivermectin for the treatment of COVID-19 may reduce the duration of illness. International Journal of Infectious Diseases. 2021;103:214-216. DOI: 10.1016/j.ijid.2020.11.191
  12. 12. Caly L, Druce JD, Catton MG, Jans DA, Wagstaff KM. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antiviral Research. 2020;178:104787. DOI: 10.1016/j.antiviral.2020.104787
  13. 13. Sharun K, Tiwari R, Dhama J, Dhama K. Dexamethasone to combat cytokine storm in COVID-19: Clinical trials and preliminary evidence. International Journal of Surgery (London, England). 2020;82:179. DOI: 10.1016/J.IJSU.2020.08.038
  14. 14. WHO. Coronavirus Disease (COVID-19): Dexamethasone. Geneva: World Health Organization; 2020. Available from: https://www.who.int/news-room/q-a-detail/coronavirus-disease-covid-19-dexamethasone [Accessed: July 13, 2021]
  15. 15. Ahmed MH, Hassan A. Dexamethasone for the treatment of coronavirus disease (COVID-19): A review. SN Comprehensive Clinical Medicine. 2020;2(12):2637-2646. DOI: 10.1007/s42399-020-00610-8
  16. 16. Hippensteel JA, LaRiviere WB, Colbert JF, Langou t-Astri CJ, Schmidt EP. Heparin as a therapy for COVID-19: Current evidence and future possibilities. American Journal of Physiology - Lung Cellular and Molecular Physiology. 2020;319(2):L211. DOI: 10.1152/AJPLUNG.00199.2020
  17. 17. Billett HH et al. Anticoagulation in COVID-19: Effect of enoxaparin, heparin, and Apixaban on mortality. Thrombosis and Haemostasis. 2020;120(12):1691. DOI: 10.1055/S-0040-1720978
  18. 18. Garcia Azorin D.Guidance for clinical case management of thrombosis with thrombocytopenia syndrome (TTS) following vaccination to prevent coronavirus disease (COVID-19). WHO COVID-19 Task Force. 2021
  19. 19. ten Cate H. Surviving Covid-19 with heparin? New England Journal of Medicine. 2021;385(9):845-846. DOI: 10.1056/NEJME2111151/SUPPL_FILE/NEJME2111151_DISCLOSURES.PDF
  20. 20. FDA COVID-19 Task Force. Coronavirus (COVID-19) Update: FDA Authorizes First Oral Antiviral for Treatment of COVID-19. Maryland, US: FDA; 2022. Available from: https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-authorizes-first-oral-antiviral-treatment-covid-19; [Accessed: April 21, 2022]
  21. 21. FDA COVID-19 Task Force. Coronavirus (COVID-19) Update: FDA Authorizes Additional Oral Antiviral for Treatment of COVID-19 in Certain Adults. Maryland, US: FDA; 2022. Available from: https://www.fda.gov/ news-events/press-announcements/ coronavirus-covid-19-update-fda-authorizes-additional-oral-antiviral-treatment-covid-19-certain; [Accessed: April 21, 2022]
  22. 22. Masyeni S et al. Molnupiravir: A lethal mutagenic drug against rapidly mutating SARS-CoV-2 – A narrative review. Journal of Medical Virology. 2022;94(7):3006-3016. DOI: 10.1002/jmv.27730
  23. 23. Wang Z, Yang L. In the age of omicron variant: Paxlovid raises new hopes of COVID-19 recovery. Journal of Medical Virology. 2021;94(5):1766-1767. DOI: 10.1002/JMV.27540
  24. 24. Mahase E. Covid-19: Pfizer’s paxlovid is 89% effective in patients at risk of serious illness, company reports. BMJ. 2021;375:n2713. DOI: 10.1136/BMJ.N2713
  25. 25. Cave JA, Phizackerley D. Molnupiravir: Evidence by press release. Drug and Therapeutics Bulletin. 2022;60(1):2-2. DOI: 10.1136/DTB.2021.000064
  26. 26. Fischer W et al. Molnupiravir, an Oral antiviral treatment for COVID-19. medRxiv. 2021;1:1-5. DOI: 10.1101/ 2021.06.17.21258639
  27. 27. Schäfer A et al. Therapeutic treatment with an oral prodrug of the remdesivir parental nucleoside is protective against SARS-CoV-2 pathogenesis in mice. Science Translational Medicine. 2022;14(643):1-5 DOI: 10.1126/scitranslmed.abm3410
  28. 28. Cox RM et al. Oral prodrug of remdesivir parent GS-441524 is efficacious against SARS-CoV-2 in ferrets. Nature Communications. 2021;12(1):1-11. DOI: 10.1038/s41467-021-26760-4
  29. 29. FDA COVID-19 Task Force. FDA Launches FAERS Dashboard for COVID-19 Therapies. Maryland, US: RAPS; 2022. Available from: https://www.raps.org/ news-and-articles/news-articles/2021/3/fda-launches-faers-dashboard-for-covid-19-therapie; [Accessed: March 17, 2022]
  30. 30. Veronin MA, Schumaker RP, Dixit R. The irony of MedWatch and the FAERS database: An assessment of data input errors and potential consequences. Journal of Pharmacy Technology. (SAGE Publications Sage CA: Los Angeles, CA) 2020;36(4) 164-167. DOI: 10.1177/8755122520928495
  31. 31. Yuan J et al. Pharmacotherapy management for COVID-19 and cardiac safety: A data mining approach for pharmacovigilance evidence from the FDA adverse event reporting system (FAERS). Drugs - Real World Outcomes. 2021;8(2):131-140. DOI: 10.1007/s40801-021-00229-8
  32. 32. Shan W, Hong D, Zhu J, Zhao Q. Assessment of the potential adverse events related to ribavirin-interferon combination for novel coronavirus therapy. Computational and Mathematical Methods in Medicine. 2020;2020:1-7. DOI: 10.1155/2020/1391583
  33. 33. Guo W et al. Informing selection of drugs for COVID-19 treatment through adverse events analysis. Scientific Reports. 2021;11(1):1-7. DOI: 10.1038/s41598-021-93500-5
  34. 34. Abdel-Rahman O. Factors associated with fatal coronavirus disease 2019 infections among cancer patients in the US FDA adverse event reporting system database. Future Oncology. 2021;17(36):5045-5051. DOI: 10.2217/fon-2021-0816
  35. 35. Zhao Y et al. Serious cardiovascular adverse events associated with hydroxychloroquine/chloroquine alone or with azithromycin in patients with COVID-19: A pharmacovigilance analysis of the FDA adverse event reporting system (FAERS). Drugs - Real World Outcomes. 2022;2022:1-11. DOI: 10.1007/S40801-022-00300-Y
  36. 36. Tuccori M et al. An overview of the preclinical discovery and development of bamlanivimab for the treatment of novel coronavirus infection (COVID-19): Reasons for limited clinical use and lessons for the future. Expert Opinion on Drug Discovery. 2021;16(12):1403-1414. DOI: 10.1080/17460441.2021.1960819
  37. 37. Perez J, Roustit M, Lepelley M, Revol B, Cracowski JL, Khouri C. Reported adverse drug reactions associated with the use of hydroxychloroquine and chloroquine during the COVID-19 pandemic. Annals of Internal Medicine. 2021;174(6):878-880. DOI: 10.7326/M20-7918
  38. 38. Geerts T, vander Heyden Y. In silico predictions of ADME-Tox properties: Drug absorption. Combinatorial Chemistry & High Throughput Screening. 2011;14(5):339-361
  39. 39. Bachevski D, Damevska K, Simeonovski V, Dimova M. Back to the basics: Propolis and COVID-19. Dermatologic Therapy. 2020;33(4):100-115. DOI: 10.1111/dth.13780
  40. 40. Berretta AA, Silveira MAD, Cóndor Capcha JM, de Jong D. Propolis and its potential against SARS-CoV-2 infection mechanisms and COVID-19 disease: Running title: Propolis against SARS-CoV-2 infection and COVID-19. Biomedicine and Pharmacotherapy. (Elsevier Masson SAS) 2020;131:110622. DOI: 10.1016/j.biopha.2020.110622
  41. 41. Sahlan M, al Faris MNH, Aditama R, Lischer K, Khayrani AC, Pratami DK. Molecular docking of South Sulawesi Propolis against fructose 1,6-Bisphosphatase as a type 2 diabetes mellitus drug. International Journal of Technology. 2020;11(5):910. DOI: 10.14716/ijtech.v11i5.4332
  42. 42. Güler HI, Tatar G, Yildiz O, Belduz AO. An investigation of ethanolic propolis extracts: Their potential inhibitor properties against ACE-II receptors for COVID-19 treatment by molecular docking study. ScienceOpen. 2020;1:1-5
  43. 43. Arba M et al. Virtual screening of the Indonesian medicinal plant and zinc databases for potential inhibitors of the RNA-dependent RNA polymerase (RdRp) of 2019 novel coronavirus. Indonesian Journal of Chemistry. 2020;20(6):1430. DOI: 10.22146/ijc.56120
  44. 44. Shivanika C, Kumar D, Ragunathan V, Tiwari P, Sumitha A, Devi B. Molecular docking, validation, dynamics simulations, and pharmacokinetic prediction of natural compounds against the SARS-CoV-2 main-protease. Journal of Biomolecular Structure and Dynamics. 2020;40(2):585-611. DOI: 10.1080/07391102.2020.1815584
  45. 45. Sukardiman M, Ervina MF, Pratama HP, Siswodihardjo S. The coronavirus disease 2019 main protease inhibitor from Andrographis paniculata (Burm.f) ness. Journal of Advanced Pharmaceutical Technology & Research. 2020;11(4):157. DOI: 10.4103/japtr.japtr_84_20
  46. 46. Zhang D, Wu K, Zhang X, Deng S, Peng B. In silico screening of Chinese herbal medicines with the potential to directly inhibit 2019 novel coronavirus. Journal of Integrative Medicine. 2020;18(2):152-158. DOI: 10.1016/j.joim.2020.02.005
  47. 47. Pang W et al. Chinese medical drugs for coronavirus disease 2019: A systematic review and meta-analysis. Integrative Medicine Research. 2020;9(3):100477. DOI: 10.1016/j.imr.2020.100477
  48. 48. Wijaya R, Hafidzhah M, Kharisma V, Ansori A, Parikesit A. COVID-19 In silico drug with Zingiber officinale natural product compound library targeting the Mpro protein. Makara Journal of Science. 2021;25(3):5. DOI: 10.7454/mss.v25i3.1244
  49. 49. Dibha AF et al. Biological activity of kencur (Kaempferia galanga L.) against SARS-CoV-2 main protease. International Journal of Health Sciences. 2022;6(S1):468-480. DOI: 10.53730/IJHS.V6NS1.4779
  50. 50. Ansori A et al. Bioactive compounds from Mangosteen (Garcinia mangostana L.) as an antiviral agent via dual inhibitor mechanism against SARSCoV- 2: An In silico approach. Pharmacognosy Journal. 2022;14(1):85-90. DOI: 10.5530/pj.2022.14.12
  51. 51. Shiloputra AF, Parikesit AA, Darmawan JT, Pricillia V, Turista DDR, Ansori ANM. An overview of the curcumin-based and Allicin bioactive compounds as potential treatment to SARS-CoV-2 with structural bioinformatics tools. Jurnal Teknologi Laboratorium. 2021;10(2):95-101. DOI: 10.29238/TEKNOLABJOURNAL. V10I2.291
  52. 52. Parikesit AA, Nurdiansyah R. Natural products repurposing of the H5N1-based lead compounds for the most fit inhibitors against 3C-like protease of SARS-CoV-2. Journal of Pharmacy & Pharmacognosy Research. 2021;9(5):730-745. DOI: 10.5281/zenodo.5529215
  53. 53. Adisurja GP, Parkesit AA. Virtual screening of the flavonoids compounds with the SARS-CoV-2 3C-like protease as the Lead compounds for the COVID-19. Coronaviruses. 2021;02(11):1-9. DOI: 10.2174/26 66796702666210222105547
  54. 54. Fadli A, Kusuma WA, Annisa IB, Heryanto R. Screening of potential Indonesia herbal compounds based on multi-label classification for 2019 coronavirus disease. Big Data and Cognitive Computing. 2021;5(4):75. DOI: 10.3390/bdcc5040075
  55. 55. Yu MJ. Natural product-like virtual libraries: Recursive atom-based enumeration. Journal of Chemical Information and Modeling. 2011;51(3):541-557. DOI: 10.1021/ci1002087
  56. 56. Bauer RA, Wurst JM, Tan DS. Expanding the range of ‘druggable’ targets with natural product-based libraries: An academic perspective. Current Opinion in Chemical Biology. 2010;14(3):308-314. DOI: 10.1016/j.cbpa.2010.02.001
  57. 57. Arakawa T, Yamasaki H, Ikeda K, Ejima D, Naito T, Koyama AH. Antiviral and virucidal activities of natural products. Current Medicinal Chemistry. 2009;16(20):2485-2497
  58. 58. Chemat F, Vian MA, Cravotto G. Green extraction of natural products: Concept and principles. International Journal of Molecular Sciences. 2012;13(7):8615-8627. DOI: 10.3390/ijms13078615
  59. 59. Cragg GM, Newman DJ. Natural products: A continuing source of novel drug leads. Biochimica et Biophysica Acta. 2013;1830(6):3670-3695. DOI: 10.1016/j.bbagen.2013.02.008
  60. 60. Mishra BB, Tiwari VK. Natural products: An evolving role in future drug discovery. European Journal of Medicinal Chemistry. 2011;46(10):4769-4807. DOI: 10.1016/j.ejmech.2011.07.057
  61. 61. Zhao H, Medema MH. Standardization for natural product synthetic biology. Natural Product Reports. 2016;33(8):920-924. DOI: 10.1039/C6NP00030D
  62. 62. Medema MH, Fischbach MA. Computational approaches to natural product discovery. Nature Chemical Biology. 2015;11(9):639-648. DOI: 10.1038/nchembio.1884
  63. 63. Amit Koparde A, Chandrashekar Doijad R, Shripal Magdum C. Natural products in drug discovery. In: Pharmacognosy - Medicinal Plants. London, UK: IntechOpen; 2019. DOI: 10.5772/intechopen.82860
  64. 64. Expósito O et al. Biotechnological production of taxol and related taxoids: Current state and prospects. Anti-Cancer Agents in Medicinal Chemistry. 2009;9(1):109-121. DOI: 10.2174/187152009787047761
  65. 65. Malik S, Cusidó RM, Mirjalili MH, Moyano E, Palazón J, Bonfill M. Production of the anticancer drug taxol in Taxus baccata suspension cultures: A review. Process Biochemistry. (Elsevier) 2011;46(1):23-34. DOI: 10.1016/j.procbio.2010.09.004
  66. 66. Pan H et al. Repurposed antiviral drugs for COVID-19 –interim WHO SOLIDARITY trial results. medRxiv. 2020;2(1):5-10. DOI: 10.1101/2020.10.15.20209817
  67. 67. Lem FF, Opook F, Lee DJH, Chee FT, Lawson FP, Chin SN. Molecular mechanism of action of repurposed drugs and traditional Chinese medicine used for the treatment of patients infected with COVID-19: A systematic scoping review. Frontiers in Pharmacology. 2021;11:2413. DOI: 10.3389/fphar.2020.585331
  68. 68. WHO (World Health Organization). Anticipating Emerging Infectious Disease Epidemics. Geneva: WHO; 2015. Available from: https://apps.who.int/iris/bitstream/handle/10665/252646/WHO-OHE-PED-2016.2-eng.pdf
  69. 69. WHO. WHO | What Is a Pandemic? Geneva: WHO; 2015. Available from: https://www.who.int/csr/disease/swineflu/frequently_asked_questions/pandemic/en/ [Accessed: December 30, 2018]
  70. 70. Yu P, Hu B, Shi ZL, Cui J. Geographical structure of bat SARS-related coronaviruses. Infection, Genetics and Evolution. (Elsevier, B.V.) 2019;69:224-229. DOI: 10.1016/j.meegid.2019.02.001
  71. 71. Delaune D et al. A novel SARS-CoV-2 related coronavirus in bats from Cambodia. Nature Communications. 2021;12(1):1-7. DOI: 10.1038/s41467-021-26809-4
  72. 72. Li LL et al. A novel SARS-CoV-2 related coronavirus with complex recombination isolated from bats in Yunnan province, China. Emerging Microbes and Infections. 2021;10(1):1683-1690. DOI: 10.1080/22221751.2021.1964925/SUPPL_FILE/TEMI_A_1964925_SM3486.DOCX
  73. 73. Zhou H et al. Identification of novel bat coronaviruses sheds light on the evolutionary origins of SARS-CoV-2 and related viruses CAS-TWAS Center of excellence for emerging infectious diseases (CEEID). bioRxiv. 2021;2(3):2021-2030 DOI: 10.1016/j. cell.2021.06.008
  74. 74. Takashita E et al. Efficacy of antiviral agents against the SARS-CoV-2 omicron subvariant BA.2. New England Journal of Medicine. 2022;386(15):1475-1477. DOI: 10.1056/NEJMC2201933/SUPPL_FILE/NEJMC2201933_DISCLOSURES.PDF
  75. 75. Pachetti M et al. Emerging SARS-CoV-2 mutation hot spots include a novel RNA-dependent-RNA polymerase variant. Journal of Translational Medicine. 2020;18(1):1-9. DOI: 10.1186/S12967-020-02344-6/FIGURES/4
  76. 76. Zeng L, Li D, Tong W, Shi T, Ning B. Biochemical features and mutations of key proteins in SARS-CoV-2 and their impacts on RNA therapeutics. Biochemical Pharmacology. 2021;189:114424. DOI: 10.1016/J.BCP.2021.114424
  77. 77. Cao Y et al. Omicron escapes the majority of existing SARS-CoV-2 neutralizing antibodies. Nature. 2021;602(7898):657-663. DOI: 10.1038/s41586-021-04385-3
  78. 78. Tambunan USF, Fardiansyah Nasution MA, Alkaff AH. Introductory chapter: Application of peptides in biomedical sciences. In: Polypeptide - New Insight into Drug Discovery and Development. London, UK: Intechopen; 2018. DOI: 10.5772/ INTECHOPEN.79297
  79. 79. Wang C et al. A human monoclonal 1 antibody blocking SARS-CoV-2 infection. bioRxiv. 2020;3(1):22-32. DOI: 10.1101/2020.03.11.987958
  80. 80. Shanmugaraj B, Siriwattananon K, Wangkanont K, Phoolcharoen W. Perspectives on monoclonal antibody therapy as potential therapeutic intervention for coronavirus disease-19 (COVID-19). Asian Pacific Journal of Allergy and Immunology. (NLM (Medline)) 2020;38(1):10-18. DOI: 10.12932/AP-200220-0773
  81. 81. Fakih TM. ARTICLE HISTORY Dermaseptin-based antiviral peptides to prevent COVID-19 through In silico molecular docking studies against SARS-CoV-2 spike protein. Pharmaceutical Sciences and Research (PSR). 2020;7(4):65-70. Available from: http://www.rcsb.org/pdb
  82. 82. Khaitov M et al. Silencing of SARS-CoV-2 with modified siRNA-peptide dendrimer formulation. Allergy. 2021;00:1-15. DOI: 10.1111/ALL.14850
  83. 83. Åkerström S, Mirazimi A, Tan YJ. Inhibition of SARS-CoV replication cycle by small interference RNAs silencing specific SARS proteins, 7a/7b, 3a/3b and S. Antiviral Research. 2007;73(3):219-227. DOI: 10.1016/j.antiviral.2006.10.008
  84. 84. Chowdhury UF, Sharif Shohan MU, Hoque KI, Beg MA, Sharif Siam MK, Moni MA. A computational approach to design potential siRNA molecules as a prospective tool for silencing nucleocapsid phosphoprotein and surface glycoprotein gene of SARS-CoV-2. Genomics. 2021;113(1):331-343. DOI: 10.1016/j.ygeno.2020.12.021

Written By

Arli Aditya Parikesit and Rizky Nurdiansyah

Published: 07 December 2022