Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Aug 15.
Published in final edited form as: Bioorg Med Chem Lett. 2021 Jun 15;46:128171. doi: 10.1016/j.bmcl.2021.128171

Synthesis and Biological Activity of Conformationally Restricted Indole-based Inhibitors of Neurotropic Alphavirus Replication: Generation of a Three-Dimensional Pharmacophore

Scott J Barraza , Janice A Sindac , Craig J Dobry +, Philip C Delekta +, Pil H Lee †,, David J Miller +, Scott D Larsen †,
PMCID: PMC8272561  NIHMSID: NIHMS1715713  PMID: 34098081

Abstract

We have previously reported the development of indole-based CNS-active antivirals for the treatment of neurotropic alphavirus infection, but further optimization is impeded by a lack of knowledge of the molecular target and binding site. Herein we describe the design, synthesis, and evaluation of a series of conformationally restricted analogues with the dual objectives of improving potency/selectivity and identifying the most bioactive conformation. Although this campaign was only modestly successful at improving potency, the sharply defined SAR of the rigid analogs enabled the definition of a three-dimensional pharmacophore, which we believe will be of value in further analog design and virtual screening for alternative antiviral leads.

Graphical Abstract

graphic file with name nihms-1715713-f0001.jpg


Neurotropic alphaviruses are NIAID Category B Priority pathogens that infect the central nervous system (CNS) and have the potential to cause severe and debilitating disease in humans and animals.1 Among these, the mosquito-borne equine encephalitis viruses (Eastern, Western, and Venezuelan) are endemic to the Americas and constitute an emergent public health risk due to the potential for human mortality2 and neurological damage,35 in addition to disruption of agriculture.67 Despite this risk, prophylactic therapies are inadequate; current vaccines lack wide availability and exhibit limited efficacy in humans,89 and recent vaccine candidates have demonstrated protection in animal models only.1013 Options for treating infection with small molecule drugs are similarly minimal and are unable to compensate for vaccine deficiencies. For example, the efficacy of the current standard of treatment, ribavirin, is constrained by poor CNS exposure.1416 Remarkably, equine encephalitis viruses have received scant historical attention to address these shortcomings.17 Only a handful of reports have been published in recent years detailing the discovery of drug-like small molecules1820 and natural products,2122 so there remains an urgent need to identify and develop effective therapies that can intercede with infection.

We have previously reported the discovery23 and optimization2425 of indole-based alphavirus RNA replication inhibitors, e.g. 1 and 2 (Figure 1), and, more recently, anthranilamide- based inhibitors possessing improved pharmacokinetic properties.26 Unfortunately, the molecular target remains elusive − a consequence of the phenotypic nature of our primary antiviral assays − and thus the architecture of the binding site that could guide design of more potent inhibitors is unknown. An approach that is often effective in such a scenario is the synthesis and evaluation of rigid analogues.2730 Of the multiple conformations that a drug can adopt, only a small subset are expected to have maximal activity if the binding site is well defined.31 It can therefore be informative to restrict lead molecules into specific conformations in an effort to pinpoint the most active ones.32 In addition to the obvious benefit of potentially defining a 3-dimensional (3D) pharmacophore, this strategy can also improve both potency and selectivity versus off-target effects.3335

Figure 1.

Figure 1.

Indole-based alphavirus replication inhibitor leads.

In the present work, we thus pursued two central objectives: (1) to conformationally restrict rotatable bonds in our leads 1 and 2, and (2) to develop a 3D pharmacophore model based on the resulting SAR that could expedite future drug design. Toward this end, we describe three different approaches to conformational restriction: 1) replacement of small functionality with sterically demanding functionality; 2) bridging of nonadjacent atoms; and 3) freezing of ring conformations. We also strove to minimize the number of additional atoms used in locking conformations because higher molecular weight could impede CNS permeability.

Analogs were synthesized according to one of two general procedures (Scheme 1). In all cases where chiral centers were present, the analogs were prepared and tested as racemates. General Procedure A was used to incorporate methyl groups onto the indole and to modify the benzyl group of lead 1. N-(4-Chlorobenzyl)indole-2-carboxylic acids 30a-c36 were condensed with ethyl isonipecotate or ethyl nipecotate and the respective esters were subsequently hydrolyzed to afford carboxylic acid intermediates 31a−c. These acids were condensed with a variety of amines (see Supporting Information) to produce analogs 316 (Tables 1 and 2).

Scheme 1.

Scheme 1.

General Procedure A reagents and conditions: (a) ethyl isonipecotate or ethyl nipecotate, EDC, HOBT, TEA, DCM, r.t.; (b) NaOH, H2O, THF, r.t.; (c) R3R4NH, EDC, HOBT, TEA, DCM, r.t.; General Procedure B reagents and conditions: (d) various reactions − see Scheme 2 and SI; (e) 30a, EDC, TEA, HOBT, DCM, r.t. PG = protecting group; G = reactive group.

Table 1.

Modification of the indole of 1.

graphic file with name nihms-1715713-t0006.jpg
No R1 R2 R3 IC50 (μM)a CC50 (μM)b
125 H H graphic file with name nihms-1715713-t0007.jpg 15.6 ± 1.8 >100
3 CH3 H graphic file with name nihms-1715713-t0008.jpg 61 ± 39 >100
4 H CH3 graphic file with name nihms-1715713-t0009.jpg >100 >100
a

Inhibition of luciferase expression in WEEV replicon assay.

b

Cell viability determined by the MTT reduction assay. Values are mean of at least n=3 independent experiments.

Table 2.

Modification of the benzyl amide of 1.

graphic file with name nihms-1715713-t0010.jpg
No R IC50 (μM)a CC50 (μM)b
125 graphic file with name nihms-1715713-t0011.jpg 15.6 ±1.8 >100
5c graphic file with name nihms-1715713-t0012.jpg 30.6 ± 7.3 >100
6 graphic file with name nihms-1715713-t0014.jpg >100 >100
7 graphic file with name nihms-1715713-t0015.jpg >100 >100
8 graphic file with name nihms-1715713-t0016.jpg >100 >100
9 graphic file with name nihms-1715713-t0017.jpg >100 >100
10 graphic file with name nihms-1715713-t0018.jpg >100 >100
11 graphic file with name nihms-1715713-t0019.jpg >100 >100
12 graphic file with name nihms-1715713-t0020.jpg 0.53 ±0.04 >100
13 graphic file with name nihms-1715713-t0021.jpg 1.7 ± 0.1 >100
14 graphic file with name nihms-1715713-t0022.jpg 7.1 ± 0.9 >100
1525 graphic file with name nihms-1715713-t0023.jpg 15.2 ± 4.7 62 ± 13
16 graphic file with name nihms-1715713-t0024.jpg >100 >50
a

Inhibition of luciferase expression in WEEV replicon assay.

b

Cell viability determined by the MTT reduction assay. Values are mean of at least n=3 independent experiments.

c

Based on nipecotic acid central ring (racemate).25

Alternatively, General Procedure B was employed to explore changes to the piperidine and amide moieties of lead 2 (Scheme 1). Piperidines, piperazines, and bicyclic amines were functionalized through a variety of reactions to afford intermediates with the general structure 34 (Scheme 2 and Supp. Info). Following deprotection, these intermediates were condensed with N- (4-chlorobenzyl)indole-2-carboxylic acid 30a to afford analogs 1729 (Table 3).

Scheme 2.

Scheme 2.

Reagents and conditions: (a) EDC, HOBT, Et3N, DCM, r.t.; (b) HCl, dioxane, r.t.; (c) COCl2, Et3N, toluene, DCM, 0 °C; (d) Na0, naphthalene, THF, r.t.; (e) PhLi, t-BuOH, LiBr, THF, −78 °C-r.t.; (f) n-BuLi, THF, r.t.

Table 3.

Modification of the piperidine amide of 2.

graphic file with name nihms-1715713-t0013.jpg
No R IC50 (μM)a CC50 (μM)b
224 graphic file with name nihms-1715713-t0025.jpg 0.53 ± 0.08 65.0
17 graphic file with name nihms-1715713-t0026.jpg >50 >50
18 graphic file with name nihms-1715713-t0027.jpg >50 >50
19 graphic file with name nihms-1715713-t0028.jpg 17.6c 66.7c
20 graphic file with name nihms-1715713-t0029.jpg 24.7 ± 3.5 77.3
21 graphic file with name nihms-1715713-t0030.jpg 24.4 ± 4.0 78.0
22 graphic file with name nihms-1715713-t0031.jpg 3.9 ± 0.4 74.2
23 graphic file with name nihms-1715713-t0032.jpg >50 >50
2424 graphic file with name nihms-1715713-t0033.jpg 1.6 ± 0.8 47.9
25 graphic file with name nihms-1715713-t0034.jpg 24.4 ± 6.3 61.0
26 graphic file with name nihms-1715713-t0035.jpg 24.6 ± 1.6 >100
27 graphic file with name nihms-1715713-t0036.jpg 0.96 ± 0.06 50.9
28 graphic file with name nihms-1715713-t0037.jpg 10.9 ± 0.6 56.5
29 graphic file with name nihms-1715713-t0038.jpg 0.49 ± 0.09 41.1
a

Inhibition of luciferase expression in WEEV replicon assay.

b

Cell viability determined by the MTT reduction assay. Values are mean of at least n=3 independent experiments.

c

Value is n=1 experiment.

The majority of intermediates of general structure 34 were amides and as such were prepared by condensation of an amine with a carboxylic acid followed by acid-promoted Boc-deprotection (e.g. 37 and 42, Scheme 2; for others, see SI). Urea 40 was prepared by condensation of two different amines with phosgene and was likewise deprotected. N-Tosyl-protected spirocyclic amino acid 433739 was similarly condensed with 4-(2-aminoethyl)-pyridine 39 but proved challenging to deprotect. Samarium (II) iodide either failed to remove the tosyl under neutral conditions or afforded reductive cleavage of the amide in the presence of tertiary amines,4041 and other methods4243 were similarly unsuccessful. Only sodium naphthalenide cleanly cleaved the S-N bond to afford amine 44.

Distinct from the other intermediates, alkenes 47 and 48 were synthesized via a traditional Wittig reaction and the Schlosser modification thereof,44 respectively, from N-boc-4-formylpiperidine 45 and phosphonium bromide 4645 (Scheme 2). Although a large number of highly E- and Z-selective olefinations exist,46 this divergent approach allowed for convenient and rapid access to both alkenes from the same reactants.All new analogues were evaluated for antiviral activity in a phenotypic cell-based assay against the WEEV genome replicon as previously described.25 Analogues were also tested for cytotoxicity under similar conditions using a colorimetric MTT assay. The data appear in Tables 1–4.

As noted earlier, definition of a 3D pharmacophore was our primary goal through the evaluation of conformationally-restricted analogs. Our initial efforts focused on the restriction of the indole substituents of the lead 125 (Table 1). Utilizing a non-covalent steric approach, two compounds were designed on the notion that strategically placed methyl groups would impede bond rotations of adjacent sidechains with minimal impact on molecular weight. Both the 7- methylindole 3 and the 3-methylindole 4 possessed substantially reduced potency, indicating that the methyl groups may be preventing rotatable access of the N-benzyl group and the 2-carboxamide to active conformations.

We next turned to restricting the flexibility of the N-benzyl amide on the isonipecotic acid frame of 1 (Table 2). Previously prepared analogs25 5 and 15 are included in the table for comparison. Remarkably, the majority of new analogs (6 − 11) exhibited no activity whatsoever, suggesting that the binding pocket for the N-benzyl amide is well defined and intolerant of many rigid conformations. One compound (12), however, proved to possess markedly better potency than all others in this series, which we do not believe can be attributed to simple increased lipophilicity or chain extension because analogs of similar ClogP (14) and length (1525 and 16) were less potent (Table 2). Interestingly, the racemic methyl analog 13 was also quite active; however its lower potency relative to 12 is further evidence that the amide binding site is well defined and not tolerant of even modest additional bulk or altered conformation. Importantly, the increased potency of 12 has apparently been achieved without a commensurate increase in cytotoxicity vs 1. Finally, the dramatic loss of potency of analogs 7 and 8, each a single methylene shorter than the corresponding active analogs 12 and 14, respectively, suggests that some degree of flexibility is required to engage the binding site. Overall, the results in Table 2 are consistent with an amide binding pocket in the unknown molecular target that is very well defined and discriminating.

Our attention then turned towards the central piperidine ring and the piperidine-4-carboxamide. Compound 224 was employed as the lead for this series due to its equipotency with 12 and greater ease of analog synthesis. Biological results are summarized in Table 3. The carboxamide imparts significant conformational bias to the linker between the piperidine and the pyridine, so we initially explored replacing it with E or Z olefins 17 and 18, respectively, which are conformationally locked bioisosteres for the two possible rotamers of the carboxamide.47 Remarkably, both analogs lost all activity, indicating that the caboxamide itself is likely making at least one key binding interaction. To explore this further, inverse amide 19 and shifted amide 20 were prepared, each proving to be over 30-fold less active than 2. This supported that a carboxamide bound directly to the piperidine 4-carbon through the carbonyl is a critical part of the pharmacophore. We therefore retained this motif through the remainder of our investigation.

Urea analog 21 and unsaturated amide 22 were examined next, each of which retained all of the atoms of the carboxamide and its position relative to the piperidine, but would be expected to have some minor conformational differences. The simple unsaturated amide retained more activity than the urea, but still lost nearly 10-fold potency. Methyl analogs 23 and 24 were particularly informative. While the previously prepared N-methyl analog 2424 only lost 2-fold potency, suggesting that the N-H is not functioning as a hydrogen bond donor, the new α-methyl analog 23 lost all activity, indicating that the carbonyl likely plays a much more significant role. The α-methyl group is either occluding the amide carbonyl from binding (sterically or conformationally) or is impeding favorable chair conformations of the piperidine. Finally, since we had shown with 24 that the N-H was not critical for activity, we prepared the highly rigid bicyclic analog 25. This analog lost most potency, obviously indicating that it is not locked in the optimal conformation. Collectively, the results for 1725 highlight the importance of a sterically unhindered and flexible carboxamide to the pharmacophore.

For the next three analogs 2628, we retained the “flexible” carboxamide moiety in the linker, and focused on freezing the conformational mobility of the piperidine ring. While the exo-bicycle 26 and the spirocycle 28 lost over 20-fold potency, the tricyclic analog 27 retained almost all of the potency of lead 2, suggesting that the locked equatorial-equatorial relationship between the 1- and 4-substituents may reflect the unknown bioactive conformation, Unfortunately, no activity improvement was realized by this conformational restraint.

Conversely, we examined an azapane analog 29 that we expected would be more flexible than piperidine 2 but about the same overall length. Interestingly, this compound was equally potent with the lead despite being a racemate. Depending on the eudismic ratio (not determined), the active enantiomer could potentially be two-fold more active, marking this compound as the most active compound to-date in the WEEV replicon assay. The simplest explanation for this positive result is that a larger population of azapane conformers are available that are favorable to binding than are available to the piperidine.

The replicon activity of azapane 29 was confirmed in an antiviral assay in FMV-infected BE(2)-C cells. At 5 μM, 29 demonstrated a nearly two-fold improvement in infected cell viability (36.1 ± 4.0 % versus negative control 19.6 ± 4.0 %), similar to the two-fold enhancement observed for lead 2 at 25 μM. At higher concentration improvement was more modest (22.9 ± 4.9 % cell viability at 25 μM), possibly due to concentration-dependent cytotoxicity.

The primary objective of this work was to establish a 3D pharmacophore model that could facilitate future optimization and enable virtual screening to identify alternative templates. We first noted that a number of small changes in the structure resulted in large differences in biological activity, suggesting that an “activity cliff” analysis48 could be useful for identifying key pharmacophoric elements. Employing DataWarrior49, we identified pairs of compounds exhibiting both high 2D structural similarity and divergent potency and ranked them according to structure activity landscape index (SALI)50 scores (Table S1 in Supporting Information). Based on the pairs having the greatest SALI values, there appear to be three pharmacophoric features most sensitive to structural changes: the distance between the terminal aryl group and the piperidine (e.g. 16 vs 15, 12 vs 6, 12 vs 14), the flexibility of the linker to the terminal aryl group (7 vs 12, 8 vs 14) and the piperidine-4-carboxamide carbonyl on the piperidine (2 vs 23, 2 vs 17/18, 2 vs 20, 2 vs 19).

In order to generate a hypothetical 3D pharmacophore, we used the Pharmacophore Elucidation tool in MOE 2019.0102.51 The approach for Pharmacophore Elucidation is to exhaustively search for all pharmacophore queries (arrangements of molecular features such as aromatic rinds, hydrophobic groups, hydrogen bond acceptors/donors, etc.) that induce good overlay of most of the active molecules and separate actives from inactives. It was assumed that all or most of the active compounds bind to the same biological target in a similar way and can therefore be overlaid.

“Active” compounds were defined as those with IC50 < 5 μM, affording a set of 7 actives and 22 inactives. These sets were used to generate a collection of pharmacophore queries such that all or most of the active compounds satisfy the queries. The generated queries were then scored by the accuracy of separating actives and inactives. The overall accuracy = m/N where N is the number of compounds in the input database and m is the sum of the number of actives that match the query and the number of inactives that do not match the query. Queries were also scored by degree of overlap of the actives with the pharmacophoric features. The overlap is between 0 and n where n is the number of actives. The larger the overlap the better the alignment. The overlap is calculated by the heavy atom root mean square deviation among the aligned compounds.

The program generated a database of 8850 queries from our analogue set. The output database contained the conformations of the high scoring alignment induced by the query, features of each query, the number of actives that match the query, the score of the alignment, the accuracy of the query in separating actives and inactives, the accuracies of the query in matching the actives/inactives, etc. The queries were sorted by the accuracies, and the top queries were examined for the alignment of actives (see Table S2 in Supporting Information for the top 10 queries.) One query (H H H a a) was selected as the best on the aggregate basis of accuracy, overlap, and agreement with the activity cliff analysis. The selected query has 5 features: 3 hydrophobic groups and 2 hydrogen bond acceptors (defined as the postulated locations of potential hydrogen bond donors).

The overall accuracy of this query is 0.93 out of 1.0; all the actives except for one (27) match this query, all of the inactives except for one do not match this query, and the overlap of the actives is 5.22 out of 6.0. The alignment of the six most active compounds overlaid on top of the pharmacophoric query is shown in Figure 2. This query contains the terminal hydrophobic group, which is consistent with the activity cliff analysis above. It also contains two hydrogen bond acceptors, one of which is the piperidine-4-carboxamide which was also shown to be important in the activity cliff analysis. A detailed description of how the pharmacophore elucidation was performed is provided in the Supporting Information.

Figure 2.

Figure 2.

The overlay of the six active analogs with the pharmacophoric query. Green represents location of hydrophobic groups and cyan indicates hydrogen bond acceptors (represented by optimal location of potential hydrogen bond donors in binding site).

In conclusion, we have explored a variety of conformationally restricted analogs of our WEEV antiviral leads 1 and 2, with several objectives in mind: 1) improving antiviral potency; 2) increasing selectivity vs off-targets that could reduce cytotoxicity; and 3) beginning to define a 3D pharmacophore for the unknown molecular target to facilitate future design, including virtual screening to potentially identify new lead chemotypes. Both covalent and non-covalent approaches to restricting rotational freedom were examined. As expected, many rigid analogs completely lost activity, suggesting that the binding site is well defined and highly discriminating based on molecular shape. Significant improvements in antiviral potency were realized with terminal amide analogs of 1 (e.g. 12) without increasing cytotoxicity, suggesting some improvement in selectivity for the unknown viral target. Modification of the carboxamide and piperidine of more potent lead 2 were less successful at improving potency but helped to identify important pharmacophoric elements. The piperidine ring had little tolerance for conformational rigidity, with only the locked 1,4-diequatorial analog 27 retaining most of the potency of lead 2. Conversely, increasing flexibility, as in azapane analog 29, actually improved potency slightly. We applied computational modeling to identify the most important SAR (activity cliffs) and to subsequently define a common pharmacophore which rationalized most of the SAR. Key elements of this pharmacophore include an optimum linker length between the indole and the terminal aromatic group of the amide N-substituent, as well as the requirement for a carboxamide with precise placement on the linker and an unhindered carbonyl. We believe this pharmacophore could be useful for designing new compounds and may enable virtual screening to identify novel viral replication inhibitors.

Supplementary Material

1

Acknowledgements

This work was supported by an NIH Partnerships for Biodefense Viral Pathogens grant (R01 AI089417, Principal Investigator DJM) and a UM Rackham Merit Scholarship for SJB.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Smith DR; Schmaljohn CS; Badger C; Ostrowski K; Zeng X; Grimes SD; Rayner JO, Comparative pathology study of Venezuelan, eastern, and western equine encephalitis viruses in nonhuman primates. Antiviral Res 2020, 182, 104875. [DOI] [PubMed] [Google Scholar]
  • 2.Zacks MA; Paessler S, Encephalitic alphaviruses. Veterinary Microbiology 2010, 140 (3−4), 281–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Feemster RF, Equine Encephalitis in Massachusetts. New Engl J Med 1957, 257 (15), 701–704. [DOI] [PubMed] [Google Scholar]
  • 4.Deresiewicz RL; Thaler SJ; Hsu LG; Zamani AA, Clinical and neuroradiographic manifestations of Eastern equine encephalitis. New Engl J Med 1997, 336 (26), 1867–1874. [DOI] [PubMed] [Google Scholar]
  • 5.Bantle CM; Phillips AT; Smeyne RJ; Rocha SM; Olson KE; Tjalkens RB, Infection with mosquito-borne alphavirus induces selective loss of dopaminergic neurons, neuroinflammation and widespread protein aggregation. NPJ Parkinsons Dis 2019, 5, 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rivas F; Diaz LA; Cardenas VM; Daza E; Bruzon L; Alcala A; De La Hoz O; Caceres FM; Aristizabal G; Martinez JW; Revelo D; De La Hoz F; Boshell J; Camacho T; Calderon L; Olano VA; Villarreal LI; Roselli D; Alvarez G; Ludwig G; Tsai T, Epidemic Venezuelan equine encephalitis in La Guajira, Colombia, 1995. Journal of Infectious Diseases 1997, 175 (4), 828–832. [DOI] [PubMed] [Google Scholar]
  • 7.Ehrenkranz NJ; Ventura AK, Venezuelan Equine Encephalitis Virus Infection in Man. 1974, 9–14. [DOI] [PubMed] [Google Scholar]
  • 8.Griffin DE, Neurotropic Alphaviruses. In Neurotropic Viral Infections, Reiss CS, Ed. Cambridge University Press: United Kingdom, 2008; pp 94–119. [Google Scholar]
  • 9.Steele KER, D.S.; Glass PJ; Hart MK; Ludwig GV; Pratt WD; Parker MD; Smith JF, Alphavirus Encephalitides. In Medical Aspects of Biological Warfare, Dembek ZF, Ed. Office of the Surgeon General: United States of America, 2007; pp 241–270. [Google Scholar]
  • 10.Dupuy LC; Richards MJ; Livingston BD; Hannaman D; Schmaljohn CS, A Multiagent Alphavirus DNA Vaccine Delivered by Intramuscular Electroporation Elicits Robust and Durable Virus-Specific Immune Responses in Mice and Rabbits and Completely Protects Mice against Lethal Venezuelan, Western, and Eastern Equine Encephalitis Virus Aerosol Challenges. J Immunol Res 2018, 2018, 8521060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Burke CW; Froude JW; Miethe S; Hulseweh B; Hust M; Glass PJ, Human-Like Neutralizing Antibodies Protect Mice from Aerosol Exposure with Western Equine Encephalitis Virus. Viruses 2018, 10 (4), 147/1–147/7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ko SY; Akahata W; Yang ES; Kong WP; Burke CW; Honnold SP; Nichols DK; Huang YS; Schieber GL; Carlton K; DaSilva L; Traina-Dorge V; Vanlandingham DL; Tsybovsky Y; Stephens T; Baxa U; Higgs S; Roy CJ; Glass PJ; Mascola JR; Nabel GJ; Rao SS, A virus-like particle vaccine prevents equine encephalitis virus infection in nonhuman primates. Sci Transl Med 2019, 11 (492), eaav3113. [DOI] [PubMed] [Google Scholar]
  • 13.Stromberg ZR; Fischer W; Bradfute SB; Kubicek-Sutherland JZ; Hraber P, Vaccine Advances against Venezuelan, Eastern, and Western Equine Encephalitis Viruses. Vaccines (Basel) 2020, 8 (2), 273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Colombo G; Lorenzini L; Zironi E; Galligioni V; Sonvico F; Balducci AG; Pagliuca G; Giuliani A; Calza L; Scagliarini A, Brain distribution of ribavirin after intranasal administration. Antiviral Res 2011, 92 (3), 408–14. [DOI] [PubMed] [Google Scholar]
  • 15.Snell NJC, Ribavirin - current status of a broad spectrum antiviral agent. Expert Opinion on Pharmacotherapy 2001, 2 (8), 1317–1324. [DOI] [PubMed] [Google Scholar]
  • 16.Huggins JW, Prospects for treatment of viral hemorrhagic fevers with ribavirin, a broad-spectrum antiviral drug. Rev Infect Dis 1989, 11 Suppl 4, S750–61. [DOI] [PubMed] [Google Scholar]
  • 17.Reichert E; Clase A; Bacetty A; Larsen J, Alphavirus antiviral drug development: scientific gap analysis and prospective research areas. Biosecur Bioterror 2009, 7 (4), 413–27. [DOI] [PubMed] [Google Scholar]
  • 18.Madsen C; Hooper I; Lundberg L; Shafagati N; Johnson A; Senina S; de la Fuente C; Hoover LI; Fredricksen BL; Dinman J; Jacobs JL; Kehn-Hall K, Small molecule inhibitors of Ago2 decrease Venezuelan equine encephalitis virus replication. Antiviral Res 2014, 112, 26–37. [DOI] [PubMed] [Google Scholar]
  • 19.Nguyen TH; Haese NN; Madadi N; Sarkar S; Bonin K; Streblow CE; Taft-Benz S; Tower NA; Rasmussen L; Bostwick R; Augelli-Szafran CE; Suto MJ; Morrison TE; DeFilippis V; Heise MT; Streblow DN; Pathak AK, Studies on Dibenzylamines as Inhibitors of Venezuelan Equine Encephalitis Virus. ACS Infect Dis 2019, 5 (12), 2014–2028. [DOI] [PubMed] [Google Scholar]
  • 20.Saikh KU; Morazzani EM; Piper AE; Bakken RR; Glass PJ, A small molecule inhibitor of MyD88 exhibits broad spectrum antiviral activity by up regulation of type I interferon. Antiviral Res 2020, 181, 104854. [DOI] [PubMed] [Google Scholar]
  • 21.Delekta PC; Raveh A; Larsen MJ; Schultz PJ; Tamayo-Castillo G; Sherman DH; Miller DJ, The combined use of alphavirus replicons and pseudoinfectious particles for the discovery of antivirals derived from natural products. J Biomol Screen 2015, 20 (5), 673–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sabini MC; Cariddi LN; Escobar FM; Mañas F; Comini L; Iglesias D; Larrauri M; Núñez Montoya S; Sereno J; Contigiani MS; Cantero JJ; Sabini LI, Potent inhibition of Western Equine Encephalitis virus by a fraction rich in flavonoids and phenolic acids obtained from Achyrocline satureioides. Revista Brasileira de Farmacognosia 2016, 26 (5), 571–578. [Google Scholar]
  • 23.Peng W; Peltier DC; Larsen MJ; Kirchhoff PD; Larsen SD; Neubig RR; Miller DJ, Identification of thieno[3,2-b]pyrrole derivatives as novel small molecule inhibitors of neurotropic alphaviruses. J Infect Dis 2009, 199 (7), 950–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sindac JA; Barraza SJ; Dobry CJ; Xiang J; Blakely PK; Irani DN; Keep RF; Miller DJ; Larsen SD, Optimization of Novel Indole-2-carboxamide Inhibitors of Neurotropic Alphavirus Replication. Journal of Medicinal Chemistry 2013, 56 (22), 9222–9241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sindac JA; Yestrepsky BD; Barraza SJ; Bolduc KL; Blakely PK; Keep RF; Irani DN; Miller DJ; Larsen SD, Novel Inhibitors of Neurotropic Alphavirus Replication That Improve Host Survival in a Mouse Model of Acute Viral Encephalitis. Journal of Medicinal Chemistry 2012, 55 (7), 3535–3545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Barraza SJ; Delekta PC; Sindac JA; Dobry CJ; Xiang J; Keep RF; Miller DJ; Larsen SD, Discovery of anthranilamides as a novel class of inhibitors of neurotropic alphavirus replication. Bioorganic & Medicinal Chemistry 2015, 23 (7), 1569–1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lemke TLW, D.A., Foye’s Principles of Medicinal Chemistry. Sixth ed.; Lippincott Williams and Wilkinds: USA, 2008. [Google Scholar]
  • 28.Vagner J; Qu H; Hruby VJ, Peptidomimetics, a synthetic tool of drug discovery. Current opinion in chemical biology 2008, 12 (3), 292–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lima LMA; Barreiro EJ, Bioisosterism: A useful strategy for molecular modification and drug design. Curr Med Chem 2005, 12 (1), 23–49. [DOI] [PubMed] [Google Scholar]
  • 30.Olesen PH, The use of bioisosteric groups in lead optimization. Curr Opin Drug Discov Devel 2001, 4 (4), 471–8. [PubMed] [Google Scholar]
  • 31.Taylor RD; Jewsbury PJ; Essex JW, A review of protein-small molecule docking methods. J Comput Aided Mol Des 2002, 16 (3), 151–166. [DOI] [PubMed] [Google Scholar]
  • 32.Wermuth CGM, A., The Practice of Medicinal Chemistry. Second ed.; Wermuth CG, Ed. Elsevier Inc.: USA, 2003. [Google Scholar]
  • 33.Dwoskin LP; Crooks PA, Competitive Neuronal Nicotinic Receptor Antagonists: A New Direction for Drug Discovery. Journal of Pharmacology and Experimental Therapeutics 2001, 298 (2), 395–402. [PubMed] [Google Scholar]
  • 34.Kazuta Y; Hirano K; Natsume K; Yamada S; Kimura R; Matsumoto S.-i.; Furuichi K; Matsuda A; Shuto S, Cyclopropane-Based Conformational Restriction of Histamine. (1S,2S)-2-(2- Aminoethyl)-1-(1H-imidazol-4-yl)cyclopropane, a Highly Selective Agonist for the Histamine H3 Receptor, Having a cis-Cyclopropane Structure. Journal of Medicinal Chemistry 2003, 46 (10), 1980–1988. [DOI] [PubMed] [Google Scholar]
  • 35.Willhite CC; Dawson MI, Structure-activity relationships of retinoids in developmental toxicology: IV. Planar cisoid conformational restriction. Toxicology and applied pharmacology 1990, 103 (2), 324–344. [DOI] [PubMed] [Google Scholar]
  • 36.Larsen S; Sindac J; Barraza S; Miller David J Arbovirus Inhibitors And Uses Thereof. US 2012/0252807 A1, 2012/03/19, 2012. [Google Scholar]
  • 37.Radchenko D; Grygorenko O; Komarov I, Synthesis of 2-azaspiro[3.3]heptane-derived amino acids: ornitine and GABA analogues. Amino Acids 2010, 39 (2), 515–521. [DOI] [PubMed] [Google Scholar]
  • 38.Radchenko DS; Pavlenko SO; Grygorenko OO; Volochnyuk DM; Shishkina SV; Shishkin OV; Komarov IV, Cyclobutane-Derived Diamines: Synthesis and Molecular Structure. The Journal of Organic Chemistry 2010, 75 (17), 5941–5952. [DOI] [PubMed] [Google Scholar]
  • 39.Allinger NL; Tushaus LA, Conformational Analysis. XLIII. Stereochemical Studies in the Cyclobutane Ring System1–3. The Journal of Organic Chemistry 1965, 30 (6), 1945–1951. [Google Scholar]
  • 40.Vedejs E; Lin S, Deprotection of Arenesulfonamides with Samarium iodide. The Journal of Organic Chemistry 1994, 59 (7), 1602–1603. [Google Scholar]
  • 41.Ankner T; Hilmersson G, Instantaneous Deprotection of Tosylamides and Esters with SmI2/Amine/Water. Organic Letters 2009, 11 (3), 503–506. [DOI] [PubMed] [Google Scholar]
  • 42.Sabitha G; Reddy BVS; Abraham S; Yadav JS, Deprotection of sulfonamides using iodotrimethylsilane. Tetrahedron Letters 1999, 40 (8), 1569–1570. [Google Scholar]
  • 43.Gethin DM; Simpkins NS, A new synthetic approach to the polyoxins: Concise stereoselective preparation of protected thymine polyoxin C. Tetrahedron 1997, 53 (42), 14417–14436. [Google Scholar]
  • 44.Wang Q; Deredas D; Huynh C; Schlosser M, Sequestered Alkyllithiums: Why Phenyllithium Alone is Suitable for Betaine-Ylide Generation. Chemistry - A European Journal 2003, 9 (2), 570–574. [DOI] [PubMed] [Google Scholar]
  • 45.Staab HA; Zipplies MF; Müller T; Storch M; Krieger C, Pyridinio-isoalloxazinophanes as Model Systems for Active-Site Complexes in Flavoenzymes: Syntheses, X-Ray Structure Analyses and Spectroscopic Properties. Chemische Berichte 1994, 127 (9), 1667–1680. [Google Scholar]
  • 46.Maryanoff BE; Reitz AB, The Wittig olefination reaction and modifications involving phosphoryl-stabilized carbanions. Stereochemistry, mechanism, and selected synthetic aspects. Chemical Reviews 1989, 89 (4), 863–927. [Google Scholar]
  • 47.Choudhary A; Raines RT, An evaluation of peptide-bond isosteres. Chembiochem 2011, 12 (12), 1801–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bajorath J; Peltason L; Wawer M; Guha R; Lajiness MS; Van Drie JH, Navigating structure-activity landscapes. Drug Discov Today 2009, 14 (13–14), 698–705. [DOI] [PubMed] [Google Scholar]
  • 49.DataWarrior v05.02.01. http://www.openmolecules.org/index.html.
  • 50.Guha R; Van Drie JH, Structure--activity landscape index: identifying and quantifying activity cliffs. J Chem Inf Model 2008, 48 (3), 646–58. [DOI] [PubMed] [Google Scholar]
  • 51.Molecular Operating Environment (MOE), 2019.0102; Chemical Computing Group ULC, 1010 Sherbrooke St. West, Suite #910, Montreal, QC, Canada, H3A 2R7, 2020. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES