CC BY 4.0 · Pharmaceutical Fronts 2025; 07(01): e1-e8
DOI: 10.1055/a-2523-2174
Review Article

Undeveloped Region in Target-Strategies and Potential in Antiviral Drug Discovery

Shaoqing Du
1   Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
,
Xinyong Liu
1   Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
,
Xueping Hu
2   Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao, People's Republic of China
,
Peng Zhan
1   Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
› Author Affiliations
Funding This work was supported by the National Natural Science Foundation of China (Grant Nos. 22208191, 22273049), the Development Plan for Youth Innovation Team of Shandong Province (Grant No. 2023KJ011), the Major Basic Research Project of Shandong Provincial Natural Science Foundation (Grant No. ZR2021ZD17), and the Science Foundation for Outstanding Young Scholars of Shandong Province (Grant No. ZR2020JQ31).
 

Abstract

Drug resistance is a looming threat to global health security, undermining the effectiveness of current treatments and increasing treatment failures. To address this challenge, it is necessary to explore innovative strategies by identifying new binding mechanisms and targeting previously undeveloped therapeutic avenues. This paper reviewed the potential of leveraging undeveloped domains to combat drug resistance and proposes a range of methodologies to accurately identify those specific targets. There is also an extensive review of the challenges associated with targeting undeveloped areas and strategies to effectively address them. In this process, the application of artificial intelligence (AI) can effectively improve the efficiency of drug design, while appropriate attention should be paid to the physicochemical and drug-like properties of pharmaceutical compounds in the realm of drug discovery. Given the above, focusing on these undeveloped areas could provide a promising pathway to address drug resistance; however, achieving this objective necessitates sustained investigative efforts and inventive approaches.


#

Introduction

Antiviral medications exert their therapeutic impact by precisely targeting different phases of the viral replication cycle, effectively curbing the capacity of viruses to infect host cells and proliferate. The mechanisms encompass a multifaceted approach: (a) preventing viral entry into cells; (b) disrupting viral replication by incorporating viral nucleic acids; (c) inhibiting viral enzyme activity; and (d) impeding the release of newly formed viral particles from host cells. Despite the effectiveness of these drugs, the emergence of drug resistance poses a significant concern, thereby it is crucial to explore novel antiviral therapies as well as strategic utilization of existing treatments. The phenomenon is frequently associated with mutations in target proteins that prevent the binding of the drug.[1] However, since the viral population is a mosaic of diverse genotypes, the diversity may emerge from elevated mutation rates during replication,[2] [3] the selection process for drug-resistant viruses can be rapid.

Current strategies to combat antiviral drug resistance focus on improving the efficacy and selectivity. These approaches include the modification of mechanisms of action, for example, exploring covalent drugs and proteolysis-targeting chimeras, and targeting allosteric sites.[4] However, in strategies aimed at enhancing efficacy, the development of drugs that target previously undeveloped sites may be a promising avenue to offer innovative therapeutic options capable of effectively addressing existing resistance issues.

Targeting undeveloped sites allows more specific drugs to be designed that minimize interference with normal cellular or physiological processes, thereby reducing toxic side effects. These undeveloped sites may exhibit lower mutation rates or play a critical role in the viral life cycle, making drugs that target them less likely to induce resistance.[5] [6] For example, nirmatrelvir and ensitrelvir both target the same binding site; however, their distinct binding modes result in minimal cross-resistance between them.[7] The development of first-in-class drugs that target previously undeveloped sites[8] could provide pharmaceutical researchers with a competitive advantage in the marketplace, as well as potentially substantial economic benefits. In this review, we will focus on chemotype-specific resistance to chemical inhibitors, as these mechanisms are increasingly being elucidated. We will highlight recent examples of drug-resistance analyses and innovative chemical strategies developed to effectively combat this resistance ([Fig. 1]).

Zoom Image
Fig. 1 Strategies aimed at targeting previously undeveloped regions to overcome resistance to targeted molecular therapeutics.

#

Inhibitors with Novel Binding Modes

Even within the confines of a single binding site, the mode of binding can be substantially different. This insight opens up a promising avenue for identifying compounds with novel mechanisms of action. M2 proton channel blockers of the influenza A virus (A/M2) are a category of antiviral agents, of which amantadine ([Fig. 2], compound 1) and rimantadine are notable examples that have been approved by the U.S. Food and Drug Administration (FDA).[9] The advent of high-resolution A/M2 structures has strengthened their role in rational drug design,[10] with three primary resistance mutations identified in A/M2 being V27A, L26F, and S31N.[11] [12]

Zoom Image
Fig. 2 Binding modes of compounds 1 (green) and 2 (pink) (PDB code: 2LY0).[14]

Molecular dynamics (MD) simulations have forecasted a pore radius of less than 1.4 Å for the Asn31 site, whereas, an expanded pore radius of 2.6 Å for the Ser31 position.[13] Compound 1 has high mobility within the N31 channel, hindering its binding efficacy. Thus, compound 2 ([Fig. 2]) was synthesized to block the channel blockade and exhibited enhanced antiviral efficacy against the S31N mutant compared to compound 1's activity against the wild-type (WT) M2, highlighting the potential of the compound for further development.[14] Compound 2 is securely anchored within the central cavity of the S31N mutant ([Fig. 2]). Within the Asn31–Gly34 region, spatial accommodation of the adamantane scaffold is restricted, whereas, the thiophene moiety of compound 2 was involved in favorable hydrophobic interactions. Compound 2 exhibits stronger inhibitory activity against the S31N mutant, which may be attributed to the increased flexibility of the compound to adopt various binding conformations within the interaction site as compared to compound 1.

The antiviral agent PF-00835231 is a rupintrivir derivative that specifically targets the main protease (Mpro) of severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), and has been subjected to further optimization to enhance its oral bioavailability. This led to the development of nirmatrelvir ([Fig. 3], compound 3). Ensitrelvir (4) was discovered through a combination of virtual screening and subsequent biological evaluation of a compound library, followed by optimization using a structure-based drug design approach.[15] The mutant ∆P168/A173V exhibits a significant 47.8-fold increase in resistance to compound 3 compared to the virus. However, a relatively mild 3.9-fold increase in resistance to compound 4.

Zoom Image
Fig. 3 Binding modes of compounds 3 (magenta) and 4 (green) at the active site of SARS-CoV-2 Mpro (PDB codes: 7VU6, 7SI9).[42] [43] The symbol ΔP168 demonstrated a single-residue deletion at position Pro168.

The observed disparities can be attributed to the distinct binding modes of the compounds ([Fig. 3]). Compound 3 has a V-shaped conformation within the binding pocket, while compound 4 has a T-shaped structure. Compound 3 forms covalent bonds with the active site of Mpro, whereas compound 4 interacts with Mpro through non-covalent interactions. These findings emphasize the potential for the development of alternative protease inhibitors that exhibit different modes of action and drug resistance profiles. The identification of these molecular mechanisms contributes to the development of future inhibitors, particularly compound 4, which requires a precise understanding of its molecular inhibition mechanism due to its characterization as the first non-covalent, non-peptide oral inhibitor.


#

Exploiting Novel Binding Cavity Spaces

It is a viable strategy to develop pharmaceuticals based on three-dimensional structural information of target proteins. By understanding the interactions between the drug and its target, along with identifying previously unexplored binding sites, drug molecules can be specifically designed to effectively occupy these binding sites. The advantage of this approach is its capacity to accurately target distinct binding sites, thereby enhancing both the specificity and potency of the drug.

A prevalent strategy in the design of HIV-1 reverse transcriptase inhibitors (RTIs) involves the synthesis of compounds that simultaneously occupy multiple binding sites.[16] [17] [18] In current antiretroviral therapy options, there is an urgent need for novel RTIs that provide enhanced resistance profiles and improved safety. By targeting the tolerant region II ([Fig. 4]), a series of innovative derivatives were identified. Compound 6 exhibits broad-spectrum antiviral activity and higher resistance against RTI-resistant variants compared to compound 5. Notably, it also inhibits cytochrome P450 enzyme's activity compared to compound 5 with a decreased likelihood of hERG blockade.[19]

Zoom Image
Fig. 4 Co-crystal structure of etravirine (5), compound 6, and HIV-1 RT complex (PDB code: 3MEC).[44]

In the quest for novel HIV protease inhibitors, a strategy emphasizing the expansion of the ligand-binding pocket has been employed ([Fig. 5]).[20] [21] For instance, the potency of darunavir (7), which features P1 phosphonate modifications (8), can be augmented against resistant variants by expanding the size of P1′ hydrophobic groups and incorporating diverse P2′ fragments.

Zoom Image
Fig. 5 Darunavir (7), and the binding mode of the corresponding P1 phosphonate analog GS-8374 (PDB code: 2I4W)[21] with compound 8.

The discovery of neuraminidase (NA) inhibitors that target the 150-cavity illustrates a similar drug design approach ([Fig. 6]). NAs are crucial glycoproteins located on the surface antigens,[22] and promising candidates for the development of anti-influenza virus therapeutics.[23] Among the approved NA inhibitors, oseltamivir, and zanamivir have been utilized worldwide since the influenza season of 1999 to 2000.[24] [25]

Zoom Image
Fig. 6 Oseltamivir carboxylate (9), and docking results of compound 10 (cyan) with NA (PDB code: 2HU0).[26]

NAs can be divided into two distinct groups, reflecting their phylogenetic relationships.[26] The 150-loop of group-1 displays an open conformation, with a 150-cavity in its active site, which suggests new opportunities for inhibitor design.[26] By incorporating substituents that specifically target the 150-cavity into existing NA inhibitors, novel drugs may be developed.[26] [27]

The 150-cavity has been the focus in the design of oseltamivir carboxylate ([Fig. 6], compound 9) and its derivatives, of which compound 10 is a potent and selective inhibitor of group-1 NA. Compound 10 showed the highest inhibitory activity against NAs ([Fig. 6]).[28] In addition, the H274Y enzyme showed sensitivity to compound 10 (IC50 = 0.16 μmol/L). Although the H274Y mutation is known to confer resistance to oseltamivir, compound 10 exhibits an impressive 12-fold increase in inhibitory activity against oseltamivir-resistant virus.

Optimal binding of compound 10 within the NA (N1 subtype) was ascertained through molecular docking according to a reported study.[28] As shown in [Fig. 6], the active site and the 150-cavity were occupied by compound 10. Subsequent optimization in this region led to the development of compounds with exceptional activity.[29] [30]

SARS-CoV-2 is thought to cause respiratory infections that can be fatal in severe cases. During the COVID-19 pandemic, the rise of various mutated strains has spurred scientific efforts to explore innovative treatment options to combat drug resistance to these variants. Currently, the FDA has authorized two categories of antiviral medications: Mpro inhibitors and RNA-dependent RNA polymerase inhibitors; however, resistance to both classes has been observed. In addition to these targets, papain-like protease (PLpro) inhibitors come to clinical research studies. Other targets identified for COVID-19 include spike and nucleocapsid proteins. These research advancements offer novel perspectives and potential targets for the clinical management of COVID-19, facilitating the development of effective therapeutic strategies.

The PLpro has emerged as a promising target in the pursuit of new mechanisms for antiviral therapy. Based on inhibitors 11 and 12 ([Fig. 7]), a novel covalent PLpro inhibitor (13) has been successfully designed. Co-crystal structure analysis revealed that compound 13 occupies the ubiquitin Val70 (Val70Ub) pocket. Leveraging this insight, the structure of compound 13 was optimized, leading to the development of compound 14, whose configuration effectively targets both the BL2 groove and Val70Ub ([Fig. 7]), showing inhibitory activity against resistant viral strains.[31]

Zoom Image
Fig. 7 Atomic model of the binding site of compound 14 (cyan) and compound 13 (pink) in SARS-CoV-2 PLpro (PDB code: 8UOB, 8UVM).[31]

According to the detailed structure of PLpro, the addition of oxadiazole and aryl carboxylic acid groups to compound 15 could significantly enhance the enzymatic inhibitory activity of the compound. As a result, a series of 1,2,4-oxadiazole derivatives were designed and synthesized. Among them, compound 16 ([Fig. 8]) demonstrated remarkable PLpro inhibitory activity (IC50 = 1.0 μmol/L) and antiviral efficacy (EC50 = 4.3 μmol/L), and also showed moderate oral bioavailability.[32]

Zoom Image
Fig. 8 Design strategy of compound 16 based on compound 15. Compound 15 is presented as a stick model (orange) (PDB code: 7CMD).[45]

The Mpro of SARS-CoV-2 is a promising target for the discovery of antiviral drugs. Most research has focused on the S4-S1 pocket; however, whether the S1′-S3′ pocket could be an innovative site for drug development remains unexplored. Notably, phenylalanine or tryptophan residues preferentially appeared at position S3′, while alanine preferentially appeared at position S1′ ([Fig. 9]). Liu et al suggested that peptide 17 (VKLQAIFR) showed a strong binding affinity with the S1′-S3′ pocket,[33] with a K d value of 3.89 μmol/L and was therefore selected as the template for further peptide design. Utilizing a warhead, a peptidomimetic inhibitor, designated as compound 18, was created. This compound has an antiviral EC50 value of 0.49 μmol/L and acts as an immune protector by inhibiting the antagonistic effect of the host NF-κB innate immune responses induced by SARS-CoV-2 Mpro.[33]

Zoom Image
Fig. 9 Co-crystal structure of peptide 17 (yellow) and the SARS-CoV-2 Mpro (PDB code: 8GWS).[33]

This design strategy is frequently employed in the development of small molecules. Based on a preclinical candidate small-molecule inhibitor ([Fig. 10], compound 19) and utilizing click chemistry for rapid discovery of potent inhibitors targeting the S2 pocket of the Mpro of SARS-CoV-2, a series of novel 1,2,3-triazole derivatives have been identified as potent inhibitors of Mpro with significant anti-SARS-CoV-2 activity. Among them, compound 20 ([Fig. 10]) demonstrates superior potency compared to nirmatrelvir (EC50 = 1.95 μmol/L) and similar efficacy to ensitrelvir (EC50 = 0.11 μmol/L). Notably, compound 20 shows robust antiviral activity against several SARS-CoV-2 variants, including Gamma, Delta, and Omicron (EC50 = 0.13–0.26 μmol/L), as well as HCoV-OC43, highlighting its broad-spectrum antiviral potential. Notably, compound 20 still retains antiviral activity against nirmatrelvir-resistant strains with T21I/E166V and L50F/E166V mutations in Mpro (EC50 = 0.26 and 0.15 μmol/L, respectively).[34]

Zoom Image
Fig. 10 Co-crystal structure of compounds 19 (cyan), 20 (pink), and the SARS-CoV-2 Mpro (PDB code: 9G01).[34]

#

Conclusion

Drug resistance poses a significant challenge to global public health, compromising the efficacy of existing treatments and resulting in treatment failures. This paper outlines strategies targeting undeveloped sites to combat the growing prevalence of drug resistance, including the identification of new binding modes and the focus on novel binding sites. These strategies have been extensively applied in the domain of antiviral research specifically concerning influenza A, SARS-CoV-2, and HIV-1.

While targeting undeveloped sites presents a promising strategy, it comes with certain challenges. An essential aspect is the identification of undeveloped sites. To identify these hidden allosteric sites, researchers have developed a variety of computational and experimental methods. For instance, MD simulations can sample multiple conformations of proteins, thereby revealing unexplored allosteric sites.[35] MD simulations are suitable for investigating the dynamic properties of proteins, such as conformational changes and allosteric effects. Markov state models can provide an atomic-level detailed view of biomolecules and help identify hidden allosteric sites. In the realm of experimental techniques, cysteine trapping, and room-temperature X-ray crystallography significantly advance the study of protein dynamics and allosteric mechanisms. These advancements facilitate the identification of undeveloped allosteric sites in large protein systems.[36] Moreover, the integration of artificial intelligence (AI) and machine learning technologies is gaining significant traction in drug design. The ability of AI to adeptly process and interpret vast bioinformatics data to reveal underlying patterns and correlations has improved the precision of potential drug target identification and accelerated the drug screening and design processes.[37] AI-driven target discovery technologies of in silico medicine have been extensively applied in various fields. For example, PandaOmics™, an AI biological target discovery platform, has successfully identified several previously unreported potential therapeutic targets for amyotrophic lateral sclerosis.[38] These technologies will facilitate the research and development of antiviral drugs targeting undeveloped sites.

The strategy of targeting new sites is often associated with an increased molecular weight after structural optimization, which subsequently alters the physicochemical properties and drug-likeness. Therefore, it is essential to conduct multidimensional drug-likeness optimization of lead compounds under the guidance of structural biology information. This approach should incorporate novel drug-likeness parameters for early virtual prediction or actual assessment of lead compounds. For instance, parameters such as ligand efficiency, lipophilic efficiency, and Fsp3 can be utilized.[39] [40] [41]

In summary, the study of previously undeveloped sites offers fresh insights and opportunities to combat drug resistance for antiviral strategies, underscoring their potential to play a role in the future management of viral infections, driven by ongoing advancements in biotechnology, and our evolving comprehension of biological processes.


#
#

Conflict of Interest

None declared.

  • References

  • 1 Clavel F, Hance AJ. HIV drug resistance. N Engl J Med 2004; 350 (10) 1023-1035
  • 2 Andino R, Domingo E. Viral quasispecies. Virology 2015; 479-480: 46-51
  • 3 Margeridon-Thermet S, Shulman NS, Ahmed A. et al. Ultra-deep pyrosequencing of hepatitis B virus quasispecies from nucleoside and nucleotide reverse-transcriptase inhibitor (NRTI)-treated patients and NRTI-naive patients. J Infect Dis 2009; 199 (09) 1275-1285
  • 4 Du S, Hu X, Menéndez-Arias L, Zhan P, Liu X. Target-based drug design strategies to overcome resistance to antiviral agents: opportunities and challenges. Drug Resist Updat 2024; 73: 101053
  • 5 Du J, Guo J, Kang D. et al. New techniques and strategies in drug discovery. Chin Chem Lett 2020; 31 (07) 1695-1708
  • 6 Jiang X, Yu J, Zhou Z. et al. Molecular design opportunities presented by solvent-exposed regions of target proteins. Med Res Rev 2019; 39 (06) 2194-2238
  • 7 Wang X, Wang YQ, Zhang Q, Yi CQ, Wang XQ. Research progress in cellular pharmacokinetics of nano-drug delivery systems. Yao Xue Xue Bao 2018; 53 (10) 1620-1629
  • 8 Lu L, Su S, Yang H, Jiang S. Antivirals with common targets against highly pathogenic viruses. Cell 2021; 184 (06) 1604-1620
  • 9 Wang J, Li F, Ma C. Recent progress in designing inhibitors that target the drug-resistant M2 proton channels from the influenza A viruses. Biopolymers 2015; 104 (04) 291-309
  • 10 Hong M, DeGrado WF. Structural basis for proton conduction and inhibition by the influenza M2 protein. Protein Sci 2012; 21 (11) 1620-1633
  • 11 Furuse Y, Suzuki A, Kamigaki T, Oshitani H. Evolution of the M gene of the influenza A virus in different host species: large-scale sequence analysis. Virol J 2009; 6: 67
  • 12 Furuse Y, Suzuki A, Oshitani H. Large-scale sequence analysis of M gene of influenza A viruses from different species: mechanisms for emergence and spread of amantadine resistance. Antimicrob Agents Chemother 2009; 53 (10) 4457-4463
  • 13 Gu RX, Liu LA, Wang YH, Xu Q, Wei DQ. Structural comparison of the wild-type and drug-resistant mutants of the influenza A M2 proton channel by molecular dynamics simulations. J Phys Chem B 2013; 117 (20) 6042-6051
  • 14 Wang J, Wu Y, Ma C. et al. Structure and inhibition of the drug-resistant S31N mutant of the M2 ion channel of influenza A virus. Proc Natl Acad Sci U S A 2013; 110 (04) 1315-1320
  • 15 Moghadasi SA, Heilmann E, Khalil AM. et al. Transmissible SARS-CoV-2 variants with resistance to clinical protease inhibitors. Sci Adv 2023; 9 (13) eade8778
  • 16 Kang D, Fang Z, Li Z. et al. Design, synthesis, and evaluation of thiophene[3,2-d]pyrimidine derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors with significantly improved drug resistance profiles. J Med Chem 2016; 59 (17) 7991-8007
  • 17 Bauman JD, Patel D, Dharia C. et al. Detecting allosteric sites of HIV-1 reverse transcriptase by X-ray crystallographic fragment screening. J Med Chem 2013; 56 (07) 2738-2746
  • 18 Frey KM, Bollini M, Mislak AC. et al. Crystal structures of HIV-1 reverse transcriptase with picomolar inhibitors reveal key interactions for drug design. J Am Chem Soc 2012; 134 (48) 19501-19503
  • 19 Wang Z, Sharma PP, Rathi B. et al. Escaping from flatland: Multiparameter optimization leads to the discovery of novel tetrahydropyrido[4,3-d]pyrimidine derivatives as human immunodeficiency virus-1 non-nucleoside reverse transcriptase inhibitors with superior antiviral activities against non-nucleoside reverse transcriptase inhibitor-resistant variants and favorable drug-like profiles. J Med Chem 2023; 66 (13) 8643-8665
  • 20 Lockbaum GJ, Rusere LN, Henes M. et al. HIV-1 protease inhibitors with a P1 phosphonate modification maintain potency against drug-resistant variants by increased interactions with flap residues. Eur J Med Chem 2023; 257: 115501
  • 21 Cihlar T, He GX, Liu X. et al. Suppression of HIV-1 protease inhibitor resistance by phosphonate-mediated solvent anchoring. J Mol Biol 2006; 363 (03) 635-647
  • 22 Yang J, Liu S, Du L, Jiang S. A new role of neuraminidase (NA) in the influenza virus life cycle: implication for developing NA inhibitors with novel mechanism of action. Rev Med Virol 2016; 26 (04) 242-250
  • 23 Loregian A, Mercorelli B, Nannetti G, Compagnin C, Palù G. Antiviral strategies against influenza virus: towards new therapeutic approaches. Cell Mol Life Sci 2014; 71 (19) 3659-3683
  • 24 McClellan K, Perry CM. Oseltamivir: a review of its use in influenza. Drugs 2001; 61 (02) 263-283
  • 25 Dunn CJ, Goa KL. Zanamivir: a review of its use in influenza. Drugs 1999; 58 (04) 761-784
  • 26 Russell RJ, Haire LF, Stevens DJ. et al. The structure of H5N1 avian influenza neuraminidase suggests new opportunities for drug design. Nature 2006; 443 (7107): 45-49
  • 27 Han N, Mu Y. Plasticity of 150-loop in influenza neuraminidase explored by Hamiltonian replica exchange molecular dynamics simulations. PLoS One 2013; 8 (04) e60995
  • 28 Xie Y, Xu D, Huang B. et al. Discovery of N-substituted oseltamivir derivatives as potent and selective inhibitors of H5N1 influenza neuraminidase. J Med Chem 2014; 57 (20) 8445-8458
  • 29 Zhang J, Murugan NA, Tian Y. et al. Structure-based optimization of N-substituted oseltamivir derivatives as potent anti-influenza A virus agents with significantly improved potency against oseltamivir-resistant N1–H274Y variant. J Med Chem 2018; 61 (22) 9976-9999
  • 30 Jia R, Zhang J, Bertagnin C. et al. Discovery of highly potent and selective influenza virus neuraminidase inhibitors targeting 150-cavity. Eur J Med Chem 2021; 212: 113097
  • 31 Tan B, Zhang X, Ansari A. et al. Design of a SARS-CoV-2 papain-like protease inhibitor with antiviral efficacy in a mouse model. Science 2024; 383 (6690): 1434-1440
  • 32 Qin B, Wu C, Zhao B. et al. Design, synthesis, and biological evaluation of 1,2,4-oxadiazole derivatives containing an aryl carboxylic acid moiety as potent sarbecovirus papain-like protease inhibitors. J Med Chem 2024; 67 (12) 10211-10232
  • 33 Liu M, Li J, Liu W. et al. The S1′–S3′ pocket of the SARS-CoV-2 main protease is critical for substrate selectivity and can be targeted with covalent inhibitors. Angew Chem Int Ed Engl 2023; 62 (41) e202309657
  • 34 Yang M, Lee MK, Gao S. et al. Miniaturized modular click chemistry-enabled rapid discovery of unique SARS-CoV-2 Mpro inhibitors with robust potency and drug-like profile. Adv Sci (Weinh) 2024; 11 (43) e2404884
  • 35 Zuzic L, Samsudin F, Shivgan AT. et al. Uncovering cryptic pockets in the SARS-CoV-2 spike glycoprotein. Structure 2022; 30 (08) 1062-1074.e4
  • 36 Lu S, Ji M, Ni D, Zhang J. Discovery of hidden allosteric sites as novel targets for allosteric drug design. Drug Discov Today 2018; 23 (02) 359-365
  • 37 Chen L, Fan Z, Chang J. et al. Sequence-based drug design as a concept in computational drug design. Nat Commun 2023; 14 (01) 4217
  • 38 Pun FW, Liu BHM, Long X. et al. Identification of therapeutic targets for amyotrophic lateral sclerosis using PandaOmics – An AI-enabled biological target discovery platform. Front Aging Neurosci 2022; 14: 914017
  • 39 Tanaka D, Tsuda Y, Shiyama T. et al. A practical use of ligand efficiency indices out of the fragment-based approach: ligand efficiency-guided lead identification of soluble epoxide hydrolase inhibitors. J Med Chem 2011; 54 (03) 851-857
  • 40 Kotesova S, Shenvi RA. Inner- and outer-sphere cross-coupling of high Fsp3 fragments. Acc Chem Res 2023; 56 (21) 3089-3098
  • 41 Johnson TW, Gallego RA, Edwards MP. Lipophilic efficiency as an important metric in drug design. J Med Chem 2018; 61 (15) 6401-6420
  • 42 Kneller DW, Li H, Phillips G. et al. Covalent narlaprevir- and boceprevir-derived hybrid inhibitors of SARS-CoV-2 main protease. Nat Commun 2022; 13 (01) 2268
  • 43 Unoh Y, Uehara S, Nakahara K. et al. Discovery of S-217622, a noncovalent oral SARS-CoV-2 3CL protease inhibitor clinical candidate for treating COVID-19. J Med Chem 2022; 65 (09) 6499-6512
  • 44 Lansdon EB, Brendza KM, Hung M. et al. Crystal structures of HIV-1 reverse transcriptase with etravirine (TMC125) and rilpivirine (TMC278): implications for drug design. J Med Chem 2010; 53 (10) 4295-4299
  • 45 Gao X, Qin B, Chen P. et al. Crystal structure of SARS-CoV-2 papain-like protease. Acta Pharm Sin B 2021; 11 (01) 237-245

Address for correspondence

Xinyong Liu, PhD
Cheeloo College of Medicine, Shandong University
44 West Culture Road, Jinan 250012
People's Republic of China   
Xueping Hu, PhD
Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University
72 Binhai Road, Qingdao 266237
People's Republic of China   
Peng Zhan, PhD
Cheeloo College of Medicine, Shandong University
44 West Culture Road, Jinan 250012
People's Republic of China   

Publication History

Received: 29 September 2024

Accepted: 23 January 2025

Article published online:
24 February 2025

© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany

  • References

  • 1 Clavel F, Hance AJ. HIV drug resistance. N Engl J Med 2004; 350 (10) 1023-1035
  • 2 Andino R, Domingo E. Viral quasispecies. Virology 2015; 479-480: 46-51
  • 3 Margeridon-Thermet S, Shulman NS, Ahmed A. et al. Ultra-deep pyrosequencing of hepatitis B virus quasispecies from nucleoside and nucleotide reverse-transcriptase inhibitor (NRTI)-treated patients and NRTI-naive patients. J Infect Dis 2009; 199 (09) 1275-1285
  • 4 Du S, Hu X, Menéndez-Arias L, Zhan P, Liu X. Target-based drug design strategies to overcome resistance to antiviral agents: opportunities and challenges. Drug Resist Updat 2024; 73: 101053
  • 5 Du J, Guo J, Kang D. et al. New techniques and strategies in drug discovery. Chin Chem Lett 2020; 31 (07) 1695-1708
  • 6 Jiang X, Yu J, Zhou Z. et al. Molecular design opportunities presented by solvent-exposed regions of target proteins. Med Res Rev 2019; 39 (06) 2194-2238
  • 7 Wang X, Wang YQ, Zhang Q, Yi CQ, Wang XQ. Research progress in cellular pharmacokinetics of nano-drug delivery systems. Yao Xue Xue Bao 2018; 53 (10) 1620-1629
  • 8 Lu L, Su S, Yang H, Jiang S. Antivirals with common targets against highly pathogenic viruses. Cell 2021; 184 (06) 1604-1620
  • 9 Wang J, Li F, Ma C. Recent progress in designing inhibitors that target the drug-resistant M2 proton channels from the influenza A viruses. Biopolymers 2015; 104 (04) 291-309
  • 10 Hong M, DeGrado WF. Structural basis for proton conduction and inhibition by the influenza M2 protein. Protein Sci 2012; 21 (11) 1620-1633
  • 11 Furuse Y, Suzuki A, Kamigaki T, Oshitani H. Evolution of the M gene of the influenza A virus in different host species: large-scale sequence analysis. Virol J 2009; 6: 67
  • 12 Furuse Y, Suzuki A, Oshitani H. Large-scale sequence analysis of M gene of influenza A viruses from different species: mechanisms for emergence and spread of amantadine resistance. Antimicrob Agents Chemother 2009; 53 (10) 4457-4463
  • 13 Gu RX, Liu LA, Wang YH, Xu Q, Wei DQ. Structural comparison of the wild-type and drug-resistant mutants of the influenza A M2 proton channel by molecular dynamics simulations. J Phys Chem B 2013; 117 (20) 6042-6051
  • 14 Wang J, Wu Y, Ma C. et al. Structure and inhibition of the drug-resistant S31N mutant of the M2 ion channel of influenza A virus. Proc Natl Acad Sci U S A 2013; 110 (04) 1315-1320
  • 15 Moghadasi SA, Heilmann E, Khalil AM. et al. Transmissible SARS-CoV-2 variants with resistance to clinical protease inhibitors. Sci Adv 2023; 9 (13) eade8778
  • 16 Kang D, Fang Z, Li Z. et al. Design, synthesis, and evaluation of thiophene[3,2-d]pyrimidine derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors with significantly improved drug resistance profiles. J Med Chem 2016; 59 (17) 7991-8007
  • 17 Bauman JD, Patel D, Dharia C. et al. Detecting allosteric sites of HIV-1 reverse transcriptase by X-ray crystallographic fragment screening. J Med Chem 2013; 56 (07) 2738-2746
  • 18 Frey KM, Bollini M, Mislak AC. et al. Crystal structures of HIV-1 reverse transcriptase with picomolar inhibitors reveal key interactions for drug design. J Am Chem Soc 2012; 134 (48) 19501-19503
  • 19 Wang Z, Sharma PP, Rathi B. et al. Escaping from flatland: Multiparameter optimization leads to the discovery of novel tetrahydropyrido[4,3-d]pyrimidine derivatives as human immunodeficiency virus-1 non-nucleoside reverse transcriptase inhibitors with superior antiviral activities against non-nucleoside reverse transcriptase inhibitor-resistant variants and favorable drug-like profiles. J Med Chem 2023; 66 (13) 8643-8665
  • 20 Lockbaum GJ, Rusere LN, Henes M. et al. HIV-1 protease inhibitors with a P1 phosphonate modification maintain potency against drug-resistant variants by increased interactions with flap residues. Eur J Med Chem 2023; 257: 115501
  • 21 Cihlar T, He GX, Liu X. et al. Suppression of HIV-1 protease inhibitor resistance by phosphonate-mediated solvent anchoring. J Mol Biol 2006; 363 (03) 635-647
  • 22 Yang J, Liu S, Du L, Jiang S. A new role of neuraminidase (NA) in the influenza virus life cycle: implication for developing NA inhibitors with novel mechanism of action. Rev Med Virol 2016; 26 (04) 242-250
  • 23 Loregian A, Mercorelli B, Nannetti G, Compagnin C, Palù G. Antiviral strategies against influenza virus: towards new therapeutic approaches. Cell Mol Life Sci 2014; 71 (19) 3659-3683
  • 24 McClellan K, Perry CM. Oseltamivir: a review of its use in influenza. Drugs 2001; 61 (02) 263-283
  • 25 Dunn CJ, Goa KL. Zanamivir: a review of its use in influenza. Drugs 1999; 58 (04) 761-784
  • 26 Russell RJ, Haire LF, Stevens DJ. et al. The structure of H5N1 avian influenza neuraminidase suggests new opportunities for drug design. Nature 2006; 443 (7107): 45-49
  • 27 Han N, Mu Y. Plasticity of 150-loop in influenza neuraminidase explored by Hamiltonian replica exchange molecular dynamics simulations. PLoS One 2013; 8 (04) e60995
  • 28 Xie Y, Xu D, Huang B. et al. Discovery of N-substituted oseltamivir derivatives as potent and selective inhibitors of H5N1 influenza neuraminidase. J Med Chem 2014; 57 (20) 8445-8458
  • 29 Zhang J, Murugan NA, Tian Y. et al. Structure-based optimization of N-substituted oseltamivir derivatives as potent anti-influenza A virus agents with significantly improved potency against oseltamivir-resistant N1–H274Y variant. J Med Chem 2018; 61 (22) 9976-9999
  • 30 Jia R, Zhang J, Bertagnin C. et al. Discovery of highly potent and selective influenza virus neuraminidase inhibitors targeting 150-cavity. Eur J Med Chem 2021; 212: 113097
  • 31 Tan B, Zhang X, Ansari A. et al. Design of a SARS-CoV-2 papain-like protease inhibitor with antiviral efficacy in a mouse model. Science 2024; 383 (6690): 1434-1440
  • 32 Qin B, Wu C, Zhao B. et al. Design, synthesis, and biological evaluation of 1,2,4-oxadiazole derivatives containing an aryl carboxylic acid moiety as potent sarbecovirus papain-like protease inhibitors. J Med Chem 2024; 67 (12) 10211-10232
  • 33 Liu M, Li J, Liu W. et al. The S1′–S3′ pocket of the SARS-CoV-2 main protease is critical for substrate selectivity and can be targeted with covalent inhibitors. Angew Chem Int Ed Engl 2023; 62 (41) e202309657
  • 34 Yang M, Lee MK, Gao S. et al. Miniaturized modular click chemistry-enabled rapid discovery of unique SARS-CoV-2 Mpro inhibitors with robust potency and drug-like profile. Adv Sci (Weinh) 2024; 11 (43) e2404884
  • 35 Zuzic L, Samsudin F, Shivgan AT. et al. Uncovering cryptic pockets in the SARS-CoV-2 spike glycoprotein. Structure 2022; 30 (08) 1062-1074.e4
  • 36 Lu S, Ji M, Ni D, Zhang J. Discovery of hidden allosteric sites as novel targets for allosteric drug design. Drug Discov Today 2018; 23 (02) 359-365
  • 37 Chen L, Fan Z, Chang J. et al. Sequence-based drug design as a concept in computational drug design. Nat Commun 2023; 14 (01) 4217
  • 38 Pun FW, Liu BHM, Long X. et al. Identification of therapeutic targets for amyotrophic lateral sclerosis using PandaOmics – An AI-enabled biological target discovery platform. Front Aging Neurosci 2022; 14: 914017
  • 39 Tanaka D, Tsuda Y, Shiyama T. et al. A practical use of ligand efficiency indices out of the fragment-based approach: ligand efficiency-guided lead identification of soluble epoxide hydrolase inhibitors. J Med Chem 2011; 54 (03) 851-857
  • 40 Kotesova S, Shenvi RA. Inner- and outer-sphere cross-coupling of high Fsp3 fragments. Acc Chem Res 2023; 56 (21) 3089-3098
  • 41 Johnson TW, Gallego RA, Edwards MP. Lipophilic efficiency as an important metric in drug design. J Med Chem 2018; 61 (15) 6401-6420
  • 42 Kneller DW, Li H, Phillips G. et al. Covalent narlaprevir- and boceprevir-derived hybrid inhibitors of SARS-CoV-2 main protease. Nat Commun 2022; 13 (01) 2268
  • 43 Unoh Y, Uehara S, Nakahara K. et al. Discovery of S-217622, a noncovalent oral SARS-CoV-2 3CL protease inhibitor clinical candidate for treating COVID-19. J Med Chem 2022; 65 (09) 6499-6512
  • 44 Lansdon EB, Brendza KM, Hung M. et al. Crystal structures of HIV-1 reverse transcriptase with etravirine (TMC125) and rilpivirine (TMC278): implications for drug design. J Med Chem 2010; 53 (10) 4295-4299
  • 45 Gao X, Qin B, Chen P. et al. Crystal structure of SARS-CoV-2 papain-like protease. Acta Pharm Sin B 2021; 11 (01) 237-245

Zoom Image
Fig. 1 Strategies aimed at targeting previously undeveloped regions to overcome resistance to targeted molecular therapeutics.
Zoom Image
Fig. 2 Binding modes of compounds 1 (green) and 2 (pink) (PDB code: 2LY0).[14]
Zoom Image
Fig. 3 Binding modes of compounds 3 (magenta) and 4 (green) at the active site of SARS-CoV-2 Mpro (PDB codes: 7VU6, 7SI9).[42] [43] The symbol ΔP168 demonstrated a single-residue deletion at position Pro168.
Zoom Image
Fig. 4 Co-crystal structure of etravirine (5), compound 6, and HIV-1 RT complex (PDB code: 3MEC).[44]
Zoom Image
Fig. 5 Darunavir (7), and the binding mode of the corresponding P1 phosphonate analog GS-8374 (PDB code: 2I4W)[21] with compound 8.
Zoom Image
Fig. 6 Oseltamivir carboxylate (9), and docking results of compound 10 (cyan) with NA (PDB code: 2HU0).[26]
Zoom Image
Fig. 7 Atomic model of the binding site of compound 14 (cyan) and compound 13 (pink) in SARS-CoV-2 PLpro (PDB code: 8UOB, 8UVM).[31]
Zoom Image
Fig. 8 Design strategy of compound 16 based on compound 15. Compound 15 is presented as a stick model (orange) (PDB code: 7CMD).[45]
Zoom Image
Fig. 9 Co-crystal structure of peptide 17 (yellow) and the SARS-CoV-2 Mpro (PDB code: 8GWS).[33]
Zoom Image
Fig. 10 Co-crystal structure of compounds 19 (cyan), 20 (pink), and the SARS-CoV-2 Mpro (PDB code: 9G01).[34]