InvestorsHub Logo

mick

08/03/20 8:52 PM

#2606 RE: mick #2605

$COCP #2https://stm.sciencemag.org/content/early/2020/08/03/scitranslmed.abc5332

3C-like protease inhibitors block coronavirus replication in vitro and improve survival in MERS-CoV-infected mice
View ORCID ProfileAthri D. Rathnayake1,*, View ORCID ProfileJian Zheng2,*, View ORCID ProfileYunjeong Kim3, View ORCID ProfileKrishani Dinali Perera3, Samantha Mackin2, View ORCID ProfileDavid K Meyerholz4, View ORCID ProfileMaithri M. Kashipathy5, View ORCID ProfileKevin P. Battaile6, View ORCID ProfileScott Lovell5, View ORCID ProfileStanley Perlman2,†, View ORCID ProfileWilliam C. Groutas1,† and View ORCID ProfileKyeong-Ok Chang3,†
See all authors and affiliations

Science Translational Medicine 03 Aug 2020:
eabc5332
DOI: 10.1126/scitranslmed.abc5332


The goal of this study was to evaluate the efficacy of 3CLpro inhibitors against human coronaviruses, including SARS-CoV-2, in a FRET enzyme assay and cell culture assays, as well as in a mouse model of MERS-CoV infection. Initial antiviral screening was performed with recombinant 3CLpro from SARS-CoV, MERS-CoV and SARS-CoV-2 in the FRET enzyme assay. Antiviral activity was then assessed in cultured Huh-7 cells infected with MERS-CoV and Vero E6 cells infected with SARS-CoV-2. Selected 3CLpro inhibitors were further examined using X-ray co-crystallization with MERS-CoV, SARS-CoV and SARS-CoV-2 3CLpro to elucidate the mechanism of action and identify the structural determinants of potency. Finally, two selected compounds were evaluated for in vivo efficacy in a mouse model of MERS-CoV infection (hDPP4-KI mice expressing human dipeptidylpeptidase 4 infected with a mouse-adapted MERS-CoV). Age- and sex-matched mice were randomly assigned into various groups for virus infection and treatment studies. Microscopic analysis of lung lesions was conducted in a blinded manner; other experiments were not blinded. No mice were excluded from analysis.

In vivo studies were performed in animal biosafety level 3 facilities at the University of Iowa. All experiments were conducted under protocols approved by the Institutional Animal Care and Use Committee at the University of Iowa according to guidelines set by the Association for the Assessment and Accreditation of Laboratory Animal Care and the U.S. Department of Agriculture.

The studies with MERS-CoV and SARS-CoV-2 were performed in biosafety level 3 facilities at the University of Iowa. All experiments were conducted under protocols approved by the Institutional Biosafety Committee at the University of Iowa according to guidelines set by the Biosafety in Microbiological and Biomedical Laboratories, the U.S. Department of Health and Human Services, the U.S. Public Health Service, the U.S. Centers for Disease Control and Prevention, and the National Institutes of Health.

Synthesis of 3CL protease inhibitors
Compounds 6a-k and 7a-k were readily synthesized as illustrated in Fig. 1 and are listed in Table 1 and Table S1. Briefly, the alcohol inputs were reacted with (L) leucine isocyanate methyl ester or (L) cyclohexylalanine isocyanate methyl ester to yield dipeptides 2 which were then hydrolyzed to the corresponding acids with lithium hydroxide in aqueous tetrahydrofuran. Subsequent coupling of the acids to glutamine surrogate methyl ester 8 (33, 34) furnished compounds 4. Lithium borohydride reduction yielded alcohols 5 which were then oxidized to the corresponding aldehydes 6 with Dess-Martin periodinane reagent. The bisulfite adducts 7 were generated by treatment with sodium bisulfite in aqueous ethanol and ethyl acetate (35).

Fluorescence resonance energy transfer (FRET) enzyme assay
The expression and purification of the 3CLpro of MERS-CoV, SARS-CoV and FIPV was performed by a standard method described previously by our lab (11, 19, 20). We also cloned and expressed the 3CLpro of SARS-CoV-2. The codon-optimized cDNA of full length of 3CLpro of SARS-CoV-2 (GenBank number MN908947.3) fused with sequences encoding 6 histidine at the N-terminal was synthesized by Integrated DNA (Coralville, IA). The synthesized gene was subcloned into the pET-28a(+) vector. The expression and purification of SARS-CoV-2 3CLpro were conducted following a standard procedure described by our lab (19). Briefly, stock solutions of compounds 6a-k and 7a-k were prepared in DMSO and diluted in assay buffer, which was comprised of 20 mM HEPES buffer, pH 8, containing NaCl (200 mM), EDTA (0.4 mM), glycerol (60%), and 6 mM dithiothreitol (DTT). The protease (3CLpro of MERS-CoV, SARS-CoV, SARS-CoV-2 or FIPV) was mixed with serial dilutions of each compound or with DMSO in 25 µL of assay buffer and incubated at 37°C for 30 min (MERS-CoV and FIPV) or at room temperature for 1 hour (SARS-CoV and SARS-CoV-2), followed by the addition of 25 µL of assay buffer containing substrate (FAM-SAVLQ/SG-QXL®520, AnaSpec, Fremont, CA). The substrate was derived from the cleavage sites on the viral polyproteins of SARS-CoV. Fluorescence readings were obtained using an excitation wavelength of 480 nm and an emission wavelength of 520 nm on a fluorescence microplate reader (FLx800; Biotec, Winoosk, VT) 1 hour following the addition of substrate. Relative fluorescence units (RFU) were determined by subtracting background values (substrate-containing well without protease) from the raw fluorescence values, as described previously (19). The dose-dependent FRET inhibition curves were fitted with a variable slope by using GraphPad Prism software (GraphPad, La Jolla, CA) in order to determine the IC50 values of the compounds.

Antiviral cell-based assays
Some compounds in 6a-k and 7a-k series were also investigated for their antiviral activity against the replication of MERS-CoV, FIPV or MHV-1 in Huh-7, CRFK or CCL1 cells, respectively (19). Briefly, medium containing DMSO (<0.1%) or each compound (up to 100 µM) was added to confluent cells, which were immediately infected with viruses at an MOI of 0.01. After incubation of the cells at 37°C for 24 hours, viral titers were determined with the TCID50 method (FIPV or MHV) with the CRFK or CLL1 cells or plaque assay with Vero81 cells (MERS-CoV). For SARS-CoV-2, confluent VeroE6 cells were inoculated with ~ 50-100 plaque forming units/well, and medium containing various concentrations of each compound and agar was applied to the cells. After 48-72 hours, plaques in each well were counted. EC50 values were determined by GraphPad Prism software using a variable slope (GraphPad, La Jolla, CA) (19). To confirm that these inhibitors also inhibit SARS-CoV-2 in primary human cells, differentiated human airway epithelial cells from 3 donors were used as previously described (36, 37). Two compounds (6j and 6e) were tested for their antiviral effects against SARS-CoV2. Briefly, airway epithelial cells were washed with PBS and SARS-CoV2 was inoculated at a MOI of 0.1 for 1 hour. After the inoculum was removed, media containing 6j (2 µM) or 6e (0.5 µM) was added. After 48 hours, cells were subjected to a freeze/thaw cycle, and virus titers were determined by plaque assay on Vero E6 cells.

Measurement of cytotoxicity
The cytotoxic dose for 50% cell death (CC50) for compounds 6a-k and 7a-k was determined in Huh-7, CRFK or CCL1 cells. Confluent cells grown in 96-well plates were incubated with various concentrations (1 to 100 µM) of each compound for 72 hours. Cell cytotoxicity was measured by a CytoTox 96 nonradioactive cytotoxicity assay kit (Promega, Madison, WI), and the CC50 values were calculated using a variable slope by GraphPad Prism software. The in vitro therapeutic index was calculated by dividing the CC50 by the EC50.

Protein purification, crystallization and data collection in X-ray crystallographic studies
MERS-CoV 3CLpro and SARS-CoV 3CLpro were purified as described previously (17, 19). An E. coli codon optimized construct encoding residues Ser 3264 to Phe 3568 of the orf1ab polyprotein (SARS-CoV-2 3CLpro, Genebank QHD43415.1) was cloned into a pET His6 Sumo TEV LIC cloning vector (2S-T, addgene). Expression and initial Ni-column purification was performed as described for MERS-CoV 3CLpro and SARS-CoV 3CLpro. The SUMO fusion elution fractions of SARS-CoV-2 were dialyzed against 20mM Tris pH 8.0, 100 mM NaCl and treated with TEV protease (1:10 w/w) overnight. This mixture was loaded onto 5 mL HisTrap HP column (GE Healthcare) equilibrated with 20 mM Tris pH 8.0, 100 mM NaCl and eluted with 20mM Tris pH 8.0, 100 mM NaCl, 500mM imidazole using an AKTA Pure FPLC. The flow through fractions, containing SARS-CoV-2 3CLpro without the SUMO fusion loaded onto a Superdex 75 10/300 GL size-exclusion column equilibrated with 20 mM Tris pH 8.0, 100 mM NaCl. The fractions were pooled and concentrated to 9.6 mg/mL for crystallization screening. Note that four residues from cloning (SNIG) remain at the N terminus following treatment with TEV protease.

Purified MERS-CoV 3CLpro, SARS-CoV 3CLpro and SARS-CoV-2 3CLpro in 100 mM NaCl, 20mM Tris pH 8.0 were concentrated to 10.6 mg/mL (0.3 mM), 22 mg/mL (0.64 mM) and 9.6 mg/mL (0.28 mM) respectively for crystallization screening. All crystallization experiments were setup using an NT8 drop-setting robot (Formulatrix Inc.) and UVXPO MRC (Molecular Dimensions) sitting drop vapor diffusion plates at 18°C. 100 nL of protein and 100 nL crystallization solution were dispensed and equilibrated against 50 µL of the latter. Stock solutions (100 mM) of compounds 6b, 6d, 6 g, 6h, 7i, and 7j were prepared in DMSO and complexes were generated by mixing 1 µL of the ligand (2 mM) with 49 µL (0.29 mM) of the protease and incubating on ice for 1 hour. Crystals of the MERS-CoV 3CLpro inhibitor complexes were obtained from the following conditions. Compounds 6b, 6d, 6 g and 6h: Proplex screen (Molecular Dimensions) condition E2 (8% (w/v) PEG 8000, 100 mM sodium citrate pH 5.0), compound 7i: Proplex screen (Molecular Dimensions) condition B8 (15% (w/v) PEG 4000, 100 mM sodium citrate pH 5.0, 100 mM magnesium chloride) and compound 7j: Index HT screen (Hampton Research) condition F6 (25% (w/v) PEG 3350, 100 mM Bis-Tris pH 5.5, 200 mM ammonium sulfate). Crystals of the SARS-CoV 3CLpro complex with compound 7j were obtained from the Index HT screen (Hampton Research) condition H8 (15% (w/v) PEG 3350, 100 mM magnesium formate). Crystals of the SARS-CoV-2 3CLpro complex with compound 7j were obtained in 1-2 days from the PACT HT screen (Molecular Dimensions) condition D7 (20% (w/v) PEG 6000, 100 mM Tris pH 8.0, 200 mM NaCl). Samples were transferred to a fresh drop containing 80% crystallant and 20% (v/v) PEG 200 before storing in liquid nitrogen. X-ray diffraction data were collected at the Advanced Photon Source beamline 17-ID using a Dectris Pilatus 6M (MERS-CoV 3CLpro and SARS-CoV 3CLpro) and Dectris Eiger2 X 9M (SARS-CoV-2 3CLpro) pixel array detector.

Solution and refinement of crystal structures
Intensities were integrated using XDS (38, 39) using Autoproc (40) and the Laue class analysis and data scaling were performed with Aimless (41). Structure solution was conducted by molecular replacement with Phaser (42) using a previously determined structure of MERS 3CLpro (PDB: 5WKK (17)) and SARS-CoV 3CLpro (PDB: 1Q2W (43)) SARS-CoV-2 3CLpro (PDB: 6LU7 (44)) as the search models. Structure refinement and manual model building were conducted with Phenix (45) and Coot (46), respectively. Disordered side chains were truncated to the point for which electron density could be observed. Structure validation was conducted with MolProbity (47) and figures were prepared using the CCP4MG package (48). Crystallographic data are provided in Table S2.

Therapeutic treatment in a mouse model of MERS-CoV infection
The two best compounds (6j and 6h) in the series were examined for their in vivo efficacy using 10-week old male hDPP4-KI mice infected with MERSMA-CoV (30). In the first study, animals were divided into three groups (n=5-6) and were lightly anesthetized with ketamine/xylazine and infected with 50 µl of 750 pfu MERSMA-CoV via intranasal inoculation. Compounds 6j or 6h were formulated in 10% ethanol and 90% PEG400 and given to mice from 1 to 10 dpi at 50 mg/kg/day (once per day) via intraperitoneal administration. The control mice received vehicle. Animals were weighed daily and monitored for 15 days. Animals were euthanized when an animal lost 30% of initial weight or at 15 dpi.

In the next study, treatment with compound 6j was delayed up to 3 dpi to determine the impact of delayed treatment on mouse survival. Animals were divided into five groups (n=5) and compound 6j (50 mg/kg/day, once per day) was administered to mice starting at one, two or three days after virus challenge (1, 2 or 3 dpi, respectively) until 10 dpi. Mice were monitored for weight loss and survival as described above for 15 days post virus challenge. As controls, vehicle (10% ethanol+90% PEG400) was administered equivalently to the experimental compound or animals received no treatment (untreated). The third study was conducted to assess the effects of therapeutic treatment of compound 6j in the lungs. For lung harvest and virus titration, animals were divided into three groups (n=4) of mice and compound 6j (50 mg/kg/day, once per day) or vehicle was administered to mice starting at 1 dpi until euthanasia. Animals were euthanized at 3 or 5 dpi, and lungs were removed aseptically, disassociated with a manual homogenizer in 1x PBS, briefly centrifuged, and supernatants removed. Samples were titered on Vero-81 cells as reported elsewhere (49). For lung histopathology analyses, animals were divided into two groups (n=5) and compound 6j (50 mg/kg/day, once per day) or vehicle was administered to mice starting at 1 dpi for 5 days. Mice were euthanized at 6 dpi, lungs were fixed with 10% formalin, and hematoxylin and eosin stained tissues were examined by a veterinary pathologist using the post-examination method of masking (50). Briefly, tissues were scored in an ordinal manner for edema and hyaline membrane formation using the following scale: 0, none; 1, rare (<5 alveoli); 2, <33% of lung fields; 3, 34-66% lung fields, and 4, >66% lung fields (30).

Statistical analysis
The analysis of survival curves in groups was performed using a Log-rank (Mantel-Cox) test and Gehan-Breslow-Wilcoxon test using GraphPad Prism Software (San Diego, CA). Log-transformed viral titers in the lungs, and lung edema and hyaline membrane formation in groups of mice were analyzed with multiple t tests using GraphPad Prism Software.

Supplementary Materials
stm.sciencemag.org/cgi/content/full/scitranslmed.abc5332/DC1

Materials and Methods

Fig. S1. X-ray cocrystal structures of compounds with coronavirus 3CLpro showing the Fo-Fc omit map.

Fig. S2. X-ray cocrystal structures of compounds with coronavirus 3CLpro showing hydrogen bond interactions.

Fig. S3. X-ray cocrystal structures of compounds with coronavirus 3CLpro showing electrostatic surface representation.

Fig. S4. X-ray cocrystal structures of comparative binding of 7j with the 3CLpro of MERS CoV, SARS-CoV and SARS-CoV-2.

Table S1. Compound IC50 in the FRET enzyme assay and CC50 in a cell culture assay for MERS-CoV 3CLpro

Table S2. Cocrystal structure data for compounds with the 3CLpro of MERS CoV and SARS CoV.

References (51–56)

Data File S1. Individual-level data for all figures and tables.

This is an open-access article distributed under the terms of the Creative Commons Attribution license, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

References and Notes
?K. V. Homes, Ed., Coronaviruses, (Lippincott Williams & Wilkins, Philadelphia, 2001), pp. 1187-1203.
?R. Lu, X. Zhao, J. Li, P. Niu, B. Yang, H. Wu, W. Wang, H. Song, B. Huang, N. Zhu, Y. Bi, X. Ma, F. Zhan, L. Wang, T. Hu, H. Zhou, Z. Hu, W. Zhou, L. Zhao, J. Chen, Y. Meng, J. Wang, Y. Lin, J. Yuan, Z. Xie, J. Ma, W. J. Liu, D. Wang, W. Xu, E. C. Holmes, G. F. Gao, G. Wu, W. Chen, W. Shi, W. Tan, Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding. Lancet 395, 565–574 (2020). doi:10.1016/S0140-6736(20)30251-8pmid:32007145CrossRefPubMedGoogle Scholar
?A. Zumla, J. F. Chan, E. I. Azhar, D. S. Hui, K. Y. Yuen, Coronaviruses - drug discovery and therapeutic options. Nat. Rev. Drug Discov. 15, 327–347 (2016). doi:10.1038/nrd.2015.37pmid:26868298CrossRefPubMedGoogle Scholar
A. L. Totura, S. Bavari, Broad-spectrum coronavirus antiviral drug discovery. Expert Opin. Drug Discov. 14, 397–412 (2019). doi:10.1080/17460441.2019.1581171pmid:30849247CrossRefPubMedGoogle Scholar
S. Skariyachan, S. B. Challapilli, S. Packirisamy, S. T. Kumargowda, V. S. Sridhar, Recent Aspects on the Pathogenesis Mechanism, Animal Models and Novel Therapeutic Interventions for Middle East Respiratory Syndrome Coronavirus Infections. Front. Microbiol. 10, 569 (2019). doi:10.3389/fmicb.2019.00569pmid:30984127CrossRefPubMedGoogle Scholar
J. Dyall, R. Gross, J. Kindrachuk, R. F. Johnson, G. G. Olinger Jr.., L. E. Hensley, M. B. Frieman, P. B. Jahrling, Middle East Respiratory Syndrome and Severe Acute Respiratory Syndrome: Current Therapeutic Options and Potential Targets for Novel Therapies. Drugs 77, 1935–1966 (2017). doi:10.1007/s40265-017-0830-1pmid:29143192CrossRefPubMedGoogle Scholar
?L. Zhang, D. Lin, X. Sun, U. Curth, C. Drosten, L. Sauerhering, S. Becker, K. Rox, R. Hilgenfeld, Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved a-ketoamide inhibitors. Science 368, 409–412 (2020). pmid:32198291Abstract/FREE Full TextGoogle Scholar
?D. Needle, G. T. Lountos, D. S. Waugh, Structures of the Middle East respiratory syndrome coronavirus 3C-like protease reveal insights into substrate specificity. Acta Crystallogr. D Biol. Crystallogr. 71, 1102–1111 (2015). doi:10.1107/S1399004715003521pmid:25945576CrossRefPubMedGoogle Scholar
R. Hilgenfeld, From SARS to MERS: Crystallographic studies on coronaviral proteases enable antiviral drug design. FEBS J. 281, 4085–4096 (2014). doi:10.1111/febs.12936pmid:25039866CrossRefPubMedGoogle Scholar
?A. Wu, Y. Wang, C. Zeng, X. Huang, S. Xu, C. Su, M. Wang, Y. Chen, D. Guo, Prediction and biochemical analysis of putative cleavage sites of the 3C-like protease of Middle East respiratory syndrome coronavirus. Virus Res. 208, 56–65 (2015). doi:10.1016/j.virusres.2015.05.018pmid:26036787CrossRefPubMedGoogle Scholar
?Y. Kim, V. Shivanna, S. Narayanan, A. M. Prior, S. Weerasekara, D. H. Hua, A. C. Kankanamalage, W. C. Groutas, K. O. Chang, Broad-spectrum inhibitors against 3C-like proteases of feline coronaviruses and feline caliciviruses. J. Virol. 89, 4942–4950 (2015). doi:10.1128/JVI.03688-14pmid:25694593Abstract/FREE Full TextGoogle Scholar
S. R. Mandadapu, M. R. Gunnam, K. C. Tiew, R. A. Uy, A. M. Prior, K. R. Alliston, D. H. Hua, Y. Kim, K. O. Chang, W. C. Groutas, Inhibition of norovirus 3CL protease by bisulfite adducts of transition state inhibitors. Bioorg. Med. Chem. Lett. 23, 62–65 (2013). doi:10.1016/j.bmcl.2012.11.026pmid:23218713CrossRefPubMedGoogle Scholar
Y. Kim, S. R. Mandadapu, W. C. Groutas, K. O. Chang, Potent inhibition of feline coronaviruses with peptidyl compounds targeting coronavirus 3C-like protease. Antiviral Res. 97, 161–168 (2013). doi:10.1016/j.antiviral.2012.11.005pmid:23219425CrossRefPubMedGoogle Scholar
S. R. Mandadapu, P. M. Weerawarna, M. R. Gunnam, K. R. Alliston, G. H. Lushington, Y. Kim, K. O. Chang, W. C. Groutas, Potent inhibition of norovirus 3CL protease by peptidyl a-ketoamides and a-ketoheterocycles. Bioorg. Med. Chem. Lett. 22, 4820–4826 (2012). doi:10.1016/j.bmcl.2012.05.055pmid:22698498CrossRefPubMedGoogle Scholar
?A. C. Galasiti Kankanamalage, Y. Kim, P. M. Weerawarna, R. A. Uy, V. C. Damalanka, S. R. Mandadapu, K. R. Alliston, N. Mehzabeen, K. P. Battaile, S. Lovell, K. O. Chang, W. C. Groutas, Structure-guided design and optimization of dipeptidyl inhibitors of norovirus 3CL protease. Structure-activity relationships and biochemical, X-ray crystallographic, cell-based, and in vivo studies. J. Med. Chem. 58, 3144–3155 (2015). doi:10.1021/jm5019934pmid:25761614CrossRefPubMedGoogle Scholar
A. C. Galasiti Kankanamalage, Y. Kim, A. D. Rathnayake, V. C. Damalanka, P. M. Weerawarna, S. T. Doyle, A. F. Alsoudi, D. M. P. Dissanayake, G. H. Lushington, N. Mehzabeen, K. P. Battaile, S. Lovell, K. O. Chang, W. C. Groutas, Structure-based exploration and exploitation of the S4 subsite of norovirus 3CL protease in the design of potent and permeable inhibitors. Eur. J. Med. Chem. 126, 502–516 (2017). doi:10.1016/j.ejmech.2016.11.027pmid:27914364CrossRefPubMedGoogle Scholar
?A. C. Galasiti Kankanamalage, Y. Kim, V. C. Damalanka, A. D. Rathnayake, A. R. Fehr, N. Mehzabeen, K. P. Battaile, S. Lovell, G. H. Lushington, S. Perlman, K. O. Chang, W. C. Groutas, Structure-guided design of potent and permeable inhibitors of MERS coronavirus 3CL protease that utilize a piperidine moiety as a novel design element. Eur. J. Med. Chem. 150, 334–346 (2018). doi:10.1016/j.ejmech.2018.03.004pmid:29544147CrossRefPubMedGoogle Scholar
?K. D. Perera, A. C. Galasiti Kankanamalage, A. D. Rathnayake, A. Honeyfield, W. Groutas, K. O. Chang, Y. Kim, Protease inhibitors broadly effective against feline, ferret and mink coronaviruses. Antiviral Res. 160, 79–86 (2018). doi:10.1016/j.antiviral.2018.10.015pmid:30342822CrossRefPubMedGoogle Scholar
?Y. Kim, S. Lovell, K. C. Tiew, S. R. Mandadapu, K. R. Alliston, K. P. Battaile, W. C. Groutas, K. O. Chang, Broad-spectrum antivirals against 3C or 3C-like proteases of picornaviruses, noroviruses, and coronaviruses. J. Virol. 86, 11754–11762 (2012). doi:10.1128/JVI.01348-12pmid:22915796Abstract/FREE Full TextGoogle Scholar
?Y. Kim, H. Liu, A. C. Galasiti Kankanamalage, S. Weerasekara, D. H. Hua, W. C. Groutas, K. O. Chang, N. C. Pedersen, Reversal of the Progression of Fatal Coronavirus Infection in Cats by a Broad-Spectrum Coronavirus Protease Inhibitor. PLOS Pathog. 12, e1005531 (2016). doi:10.1371/journal.ppat.1005531pmid:27027316CrossRefPubMedGoogle Scholar
?N. C. Pedersen, Y. Kim, H. Liu, A. C. Galasiti Kankanamalage, C. Eckstrand, W. C. Groutas, M. Bannasch, J. M. Meadows, K. O. Chang, Efficacy of a 3C-like protease inhibitor in treating various forms of acquired feline infectious peritonitis. J. Feline Med. Surg. 20, 378–392 (2018). doi:10.1177/1098612X17729626pmid:28901812CrossRefPubMedGoogle Scholar
?D. L. Ng, F. Al Hosani, M. K. Keating, S. I. Gerber, T. L. Jones, M. G. Metcalfe, S. Tong, Y. Tao, N. N. Alami, L. M. Haynes, M. A. Mutei, L. Abdel-Wareth, T. M. Uyeki, D. L. Swerdlow, M. Barakat, S. R. Zaki, Clinicopathologic, Immunohistochemical, and Ultrastructural Findings of a Fatal Case of Middle East Respiratory Syndrome Coronavirus Infection in the United Arab Emirates, April 2014. Am. J. Pathol. 186, 652–658 (2016). doi:10.1016/j.ajpath.2015.10.024pmid:26857507CrossRefPubMedGoogle Scholar
?T. P. Sheahan, A. C. Sims, S. R. Leist, A. Schäfer, J. Won, A. J. Brown, S. A. Montgomery, A. Hogg, D. Babusis, M. O. Clarke, J. E. Spahn, L. Bauer, S. Sellers, D. Porter, J. Y. Feng, T. Cihlar, R. Jordan, M. R. Denison, R. S. Baric, Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat. Commun. 11, 222 (2020). doi:10.1038/s41467-019-13940-6pmid:31924756CrossRefPubMedGoogle Scholar
E. de Wit, F. Feldmann, J. Cronin, R. Jordan, A. Okumura, T. Thomas, D. Scott, T. Cihlar, H. Feldmann, Prophylactic and therapeutic remdesivir (GS-5734) treatment in the rhesus macaque model of MERS-CoV infection. Proc. Natl. Acad. Sci. U.S.A. 117, 6771–6776 (2020). doi:10.1073/pnas.1922083117pmid:32054787Abstract/FREE Full TextGoogle Scholar
T. K. Warren, R. Jordan, M. K. Lo, A. S. Ray, R. L. Mackman, V. Soloveva, D. Siegel, M. Perron, R. Bannister, H. C. Hui, N. Larson, R. Strickley, J. Wells, K. S. Stuthman, S. A. Van Tongeren, N. L. Garza, G. Donnelly, A. C. Shurtleff, C. J. Retterer, D. Gharaibeh, R. Zamani, T. Kenny, B. P. Eaton, E. Grimes, L. S. Welch, L. Gomba, C. L. Wilhelmsen, D. K. Nichols, J. E. Nuss, E. R. Nagle, J. R. Kugelman, G. Palacios, E. Doerffler, S. Neville, E. Carra, M. O. Clarke, L. Zhang, W. Lew, B. Ross, Q. Wang, K. Chun, L. Wolfe, D. Babusis, Y. Park, K. M. Stray, I. Trancheva, J. Y. Feng, O. Barauskas, Y. Xu, P. Wong, M. R. Braun, M. Flint, L. K. McMullan, S. S. Chen, R. Fearns, S. Swaminathan, D. L. Mayers, C. F. Spiropoulou, W. A. Lee, S. T. Nichol, T. Cihlar, S. Bavari, Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys. Nature 531, 381–385 (2016). doi:10.1038/nature17180pmid:26934220CrossRefPubMedGoogle Scholar
?T. P. Sheahan, A. C. Sims, R. L. Graham, V. D. Menachery, L. E. Gralinski, J. B. Case, S. R. Leist, K. Pyrc, J. Y. Feng, I. Trantcheva, R. Bannister, Y. Park, D. Babusis, M. O. Clarke, R. L. Mackman, J. E. Spahn, C. A. Palmiotti, D. Siegel, A. S. Ray, T. Cihlar, R. Jordan, M. R. Denison, R. S. Baric, Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci. Transl. Med. 9, eaal3653 (2017). doi:10.1126/scitranslmed.aal3653pmid:28659436Abstract/FREE Full TextGoogle Scholar
?V. J. Munster, F. Feldmann, B. N. Williamson, N. van Doremalen, L. Pérez-Pérez, J. Schulz, K. Meade-White, A. Okumura, J. Callison, B. Brumbaugh, V. A. Avanzato, R. Rosenke, P. W. Hanley, G. Saturday, D. Scott, E. R. Fischer, E. de Wit, Respiratory disease in rhesus macaques inoculated with SARS-CoV-2. Nature (2020). doi:10.1038/s41586-020-2324-7pmid:32396922CrossRefPubMedGoogle Scholar
?T. P. Sheahan, A. C. Sims, S. Zhou, R. L. Graham, A. J. Pruijssers, M. L. Agostini, S. R. Leist, A. Schäfer, K. H. Dinnon 3rd., L. J. Stevens, J. D. Chappell, X. Lu, T. M. Hughes, A. S. George, C. S. Hill, S. A. Montgomery, A. J. Brown, G. R. Bluemling, M. G. Natchus, M. Saindane, A. A. Kolykhalov, G. Painter, J. Harcourt, A. Tamin, N. J. Thornburg, R. Swanstrom, M. R. Denison, R. S. Baric, An orally bioavailable broad-spectrum antiviral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice. Sci. Transl. Med. 12, eabb5883 (2020). doi:10.1126/scitranslmed.abb5883pmid:32253226Abstract/FREE Full TextGoogle Scholar
?A. M. Prior, Y. Kim, S. Weerasekara, M. Moroze, K. R. Alliston, R. A. Uy, W. C. Groutas, K. O. Chang, D. H. Hua, Design, synthesis, and bioevaluation of viral 3C and 3C-like protease inhibitors. Bioorg. Med. Chem. Lett. 23, 6317–6320 (2013). doi:10.1016/j.bmcl.2013.09.070pmid:24125888CrossRefPubMedGoogle Scholar
?K. Li, C. Wohlford-Lenane, S. Perlman, J. Zhao, A. K. Jewell, L. R. Reznikov, K. N. Gibson-Corley, D. K. Meyerholz, P. B. McCray Jr.., Middle East Respiratory Syndrome Coronavirus Causes Multiple Organ Damage and Lethal Disease in Mice Transgenic for Human Dipeptidyl Peptidase 4. J. Infect. Dis. 213, 712–722 (2016). pmid:26486634CrossRefPubMedGoogle Scholar
A. S. Agrawal, T. Garron, X. Tao, B. H. Peng, M. Wakamiya, T. S. Chan, R. B. Couch, C. T. Tseng, Generation of a transgenic mouse model of Middle East respiratory syndrome coronavirus infection and disease. J. Virol. 89, 3659–3670 (2015). doi:10.1128/JVI.03427-14pmid:25589660Abstract/FREE Full TextGoogle Scholar
?K. Li, C. L. Wohlford-Lenane, R. Channappanavar, J. E. Park, J. T. Earnest, T. B. Bair, A. M. Bates, K. A. Brogden, H. A. Flaherty, T. Gallagher, D. K. Meyerholz, S. Perlman, P. B. McCray Jr.., Mouse-adapted MERS coronavirus causes lethal lung disease in human DPP4 knockin mice. Proc. Natl. Acad. Sci. U.S.A. 114, E3119–E3128 (2017). doi:10.1073/pnas.1619109114pmid:28348219Abstract/FREE Full TextGoogle Scholar
?P. S. Dragovich, T. J. Prins, R. Zhou, S. E. Webber, J. T. Marakovits, S. A. Fuhrman, A. K. Patick, D. A. Matthews, C. A. Lee, C. E. Ford, B. J. Burke, P. A. Rejto, T. F. Hendrickson, T. Tuntland, E. L. Brown, J. W. Meador 3rd., R. A. Ferre, J. E. Harr, M. B. Kosa, S. T. Worland, Structure-based design, synthesis, and biological evaluation of irreversible human rhinovirus 3C protease inhibitors. 4. Incorporation of P1 lactam moieties as L-glutamine replacements. J. Med. Chem. 42, 1213–1224 (1999). doi:10.1021/jm9805384pmid:10197965CrossRefPubMedWeb of ScienceGoogle Scholar
?Q. P. Tian, N. K. Nayyar, S. Babu, L. J. Chen, J. H. Tao, S. Lee, A. Tibbetts, T. Moran, J. Liou, M. Guo, T. P. Kennedy, An efficient synthesis of a key intermediate for the preparation of the rhinovirus protease inhibitor AG7088 via asymmetric dianionic cyanomethylation of N-Boc-L-(+)-glutamic acid dimethyl ester. Tetrahedron Lett. 42, 6807–6809 (2001). doi:10.1016/S0040-4039(01)01416-2CrossRefGoogle Scholar
?D. P. Kjell, B. J. Slattery, M. J. Semo, A novel, nonaqueous method for regeneration of aldehydes from bisulfite adducts. J. Org. Chem. 64, 5722–5724 (1999). doi:10.1021/jo990543vpmid:11674650CrossRefPubMedGoogle Scholar
?P. H. Karp, T. O. Moninger, S. P. Weber, T. S. Nesselhauf, J. L. Launspach, J. Zabner, M. J. Welsh, An in vitro model of differentiated human airway epithelia. Methods for establishing primary cultures. Methods Mol. Biol. 188, 115–137 (2002). doi:10.1385/1-59259-185-X:115pmid:11987537CrossRefPubMedGoogle Scholar
?H. P. Jia, D. C. Look, L. Shi, M. Hickey, L. Pewe, J. Netland, M. Farzan, C. Wohlford-Lenane, S. Perlman, P. B. McCray Jr.., ACE2 receptor expression and severe acute respiratory syndrome coronavirus infection depend on differentiation of human airway epithelia. J. Virol. 79, 14614–14621 (2005). doi:10.1128/JVI.79.23.14614-14621.2005pmid:16282461Abstract/FREE Full TextGoogle Scholar
?W. Kabsch, Xds. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 (2010). doi:10.1107/S0907444909047337pmid:20124692CrossRefPubMedWeb of ScienceGoogle Scholar
?W. Kabsch, Automatic indexing of rotation diffraction patterns. J. Appl. Cryst. 21, 67–72 (1988). doi:10.1107/S0021889887009737CrossRefWeb of ScienceGoogle Scholar
?C. Vonrhein, C. Flensburg, P. Keller, A. Sharff, O. Smart, W. Paciorek, T. Womack, G. Bricogne, Data processing and analysis with the autoPROC toolbox. Acta Crystallogr. D Struct. Biol. 67, 293–302 (2011). doi:10.1107/S0907444911007773pmid:21460447CrossRefPubMedWeb of ScienceGoogle Scholar
?P. R. Evans, An introduction to data reduction: Space-group determination, scaling and intensity statistics. Acta Crystallogr. D Biol. Crystallogr. 67, 282–292 (2011). doi:10.1107/S090744491003982Xpmid:21460446CrossRefPubMedWeb of ScienceGoogle Scholar
?A. J. McCoy, R. W. Grosse-Kunstleve, P. D. Adams, M. D. Winn, L. C. Storoni, R. J. Read, Phaser crystallographic software. J. Appl. Cryst. 40, 658–674 (2007). doi:10.1107/S0021889807021206pmid:19461840CrossRefPubMedWeb of ScienceGoogle Scholar
?J. B. Bonanno, R. Fowler, S. Gupta, J. Hendle, D. Lorimer, R. Romero, J. M. Sauder, C. L. Wei, E. T. Liu, S. K. Burley, T. Harris, in N Y Times Web. (2003), pp. C2-C2.
?N. Zhu, D. Zhang, W. Wang, X. Li, B. Yang, J. Song, X. Zhao, B. Huang, W. Shi, R. Lu, P. Niu, F. Zhan, X. Ma, D. Wang, W. Xu, G. Wu, G. F. Gao, W. Tan; China Novel Coronavirus Investigating and Research Team, A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 382, 727–733 (2020). doi:10.1056/NEJMoa2001017pmid:31978945CrossRefPubMedGoogle Scholar
?P. V. Afonine, R. W. Grosse-Kunstleve, N. Echols, J. J. Headd, N. W. Moriarty, M. Mustyakimov, T. C. Terwilliger, A. Urzhumtsev, P. H. Zwart, P. D. Adams, Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. D Struct. Biol. 68, 352–367 (2012). doi:10.1107/S0907444912001308pmid:22505256CrossRefPubMedWeb of ScienceGoogle Scholar
?P. Emsley, B. Lohkamp, W. G. Scott, K. Cowtan, Features and development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66, 486–501 (2010). doi:10.1107/S0907444910007493pmid:20383002CrossRefPubMedWeb of ScienceGoogle Scholar
?V. B. Chen, W. B. Arendall 3rd., J. J. Headd, D. A. Keedy, R. M. Immormino, G. J. Kapral, L. W. Murray, J. S. Richardson, D. C. Richardson, MolProbity: All-atom structure validation for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 66, 12–21 (2010). doi:10.1107/S0907444909042073pmid:20057044CrossRefPubMedWeb of ScienceGoogle Scholar
?L. Potterton, S. McNicholas, E. Krissinel, J. Gruber, K. Cowtan, P. Emsley, G. N. Murshudov, S. Cohen, A. Perrakis, M. Noble, Developments in the CCP4 molecular-graphics project. Acta Crystallogr. D Struct. Biol. 60, 2288–2294 (2004). doi:10.1107/S0907444904023716pmid:15572783CrossRefPubMedWeb of ScienceGoogle Scholar
?J. Zhao, K. Li, C. Wohlford-Lenane, S. S. Agnihothram, C. Fett, J. Zhao, M. J. Gale Jr.., R. S. Baric, L. Enjuanes, T. Gallagher, P. B. McCray Jr.., S. Perlman, Rapid generation of a mouse model for Middle East respiratory syndrome. Proc. Natl. Acad. Sci. U.S.A. 111, 4970–4975 (2014). doi:10.1073/pnas.1323279111pmid:24599590Abstract/FREE Full TextGoogle Scholar
?D. K. Meyerholz, A. P. Beck, Principles and approaches for reproducible scoring of tissue stains in research. Lab. Invest. 98, 844–855 (2018). doi:10.1038/s41374-018-0057-0pmid:29849125CrossRefPubMedGoogle Scholar
?P. R. Evans, An introduction to data reduction: Space-group determination, scaling and intensity statistics. Acta Crystallogr. D Biol. Crystallogr. 67, 282–292 (2011). doi:10.1107/S090744491003982Xpmid:21460446CrossRefPubMedWeb of ScienceGoogle Scholar
P. Evans, Scaling and assessment of data quality. Acta Crystallogr. D Biol. Crystallogr. 62, 72–82 (2006). doi:10.1107/S0907444905036693pmid:16369096CrossRefPubMedWeb of ScienceGoogle Scholar
K. Diederichs, P. A. Karplus, Improved R-factors for diffraction data analysis in macromolecular crystallography. Nat. Struct. Biol. 4, 269–275 (1997). doi:10.1038/nsb0497-269pmid:9095194CrossRefPubMedWeb of ScienceGoogle Scholar
M. S. Weiss, Global indicators of X-ray data quality. J. Appl. Cryst. 34, 130–135 (2001). doi:10.1107/S0021889800018227CrossRefWeb of ScienceGoogle Scholar
P. Evans, Biochemistry. Resolving some old problems in protein crystallography. Science 336, 986–987 (2012). doi:10.1126/science.1222162pmid:22628641Abstract/FREE Full TextGoogle Scholar
?P. A. Karplus, K. Diederichs, Linking crystallographic model and data quality. Science 336, 1030–1033 (2012). doi:10.1126/science.1218231pmid:22628654Abstract/FREE Full TextGoogle Scholar
ACKNOWLEDGMENTS: The authors thank David George for technical assistance. Funding: This research was supported in part by grants from the National Institutes of Health (NIH) (R01 AI109039 to K.O.C. and P01 AI060699 and R01 AI129269 to S. P.). Use of the University of Kansas Protein Structure Laboratory was supported by a grant from the National Institute of General Medical Sciences (P30GM110761) of the NIH. Use of the IMCA-CAT beamline 17-ID at the Advanced Photon Source was supported by the companies of the Industrial Macromolecular Crystallography Association through a contract with Hauptman-Woodward Medical Research Institute. Use of the Advanced Photon Source was supported by the U.S. Department of Energy, Office of Science, Office of Basic Energy Sciences under contract no. DE-AC02-06CH11357. Author contributions: KOC, YK, SP, WCG conceptualized the study; WCG designed the compound series optimization; ADR executed compound synthesis; SP designed cell culture and animal studies; JZ and SM investigated cell culture assays with MERS-CoV and SARS-CoV2, respectively; JZ executed animal studies with selected compounds, and KDP performed histopathology; SL designed X-ray cocrystallography studies and analysis; SL, MMK and KPB performed X-ray cocrystallography. KOC and YK established enzyme assays and provided enzymes for X-ray cocrystallography; YK and KDP performed inhibitor testing in the FRET enzyme assay; KOC, YK, WCG, SL prepared the original draft of the paper; KOC, YK, WCG, SP, SL reviewed and edited the paper. Competing interests: The authors declare no competing interests. Kansas State University, Wichita State University and the University of Iowa have jointly filed a provisional patent application #63001781 entitled “Broad spectrum antivirals against coronaviruses” that covers compounds 6j and 6h as well as related compounds, with KOC, YK, SP and WCG as coinventors. Data and materials availability: All data associated with this study are in the main text or Supplementary Materials. Coronavirus 3CLpro enzymes and inhibitors are available from Kyeong-Ok Chang under a materials transfer agreement with Kansas State University. Coordinates and structure factors for the 3CLpro inhibitor complexes are deposited in the Worldwide Protein Data Bank (wwPDB) with the accession codes: MERS-CoV 3CLpro with inhibitor 6b (6VGY), 6d (6VGZ), 6 g (6VH0), 6h (6VH1), 7i (6VH2) and 7j (6VH3); SARS-CoV 3CLpro with inhibitor 7j (6W2A); SARS-CoV-2 3CLpro with inhibitor 7j (6XMK). This work is licensed under a Creative Commons Attribution 4.0 International (CC BY 4.0) license, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. To view a copy of this license, visit https://creativecommons.org/licenses/by/4.0/. This license does not apply to figures/photos/artwork or other content included in the article that is credited to a third party; obtain authorization from the rights holder before using such material.
Copyright © , American Association for the Advancement of Science
View Abstract