Abstract
We report the Synthesis, biological evaluation, and X-ray structural studies of a series of SARS-CoV-2 Mpro inhibitors based upon the X-ray structure of nirmatrelvir, an FDA approved drug that targets the main protease of SARS-CoV-2. The studies involved examination of various P4 moieties, P1 five- and six-membered lactam rings to improve potency. In particular, the six-membered P1 lactam ring analogs exhibited high SARS-CoV-2 Mpro inhibitory activity. Several compounds effectively blocked SARSCoV-2 replication in VeroE6 cells. One of these compounds maintained good antiviral activity against variants of concern including Delta and Omicron variants. A high-resolution X-ray crystal structure of an inhibitor bound to SARS-CoV-2 Mpro was determined to gain insight into the ligand-binding properties in the Mpro active site.
Keywords: Antiviral, COVID-19, Protease inhibitor, SARS-CoV-2 Mpro, Synthesis, X-ray structure
Graphical Abstract

1. Introduction
The global COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has lasted for over three years, resulting in worldwide crisis encompassing both health and economics.1,2 With nearly 7 million global deaths and 769 million cases reported to date, the outbreak is far from over. SARS-CoV-2 continues to cause a large number of severe illnesses, hospitalizations, and deaths worldwide.3,4,5 The development of COVID-19 vaccines and the large-scale vaccination efforts resulted in much needed relief from the early devastating situation.6,7 However, achieving herd immunity using vaccines has remained quite uncertain.8,9 Also, the emergence of several deadly variants caused many concerns regarding the possible return of the pandemic.10,11 Therefore, the continued development of effective antivirals is critically important to prevent COVID-related ailments, hospitalization and to prepare for potential drug-resistant SARS-CoV-2.12,13 Recently, the first orally active SARS-CoV-2 main protease (Mpro) inhibitor drug nirmatrelvir (1, Figure 1) was approved in combination with ritonavir (2), a CYP3A4 inhibitor to boost pharmacokinetic properties.14,15 Molnupiravir (3), a ribonucleoside derivative that blocks RNA-dependent RNA polymerase (RdRp) activity has also been approved recently.16,17 Both drugs have been shown to reduce the risk of hospitalization and deaths for COVID-related illnesses.18,19
Figure 1.
Structure of nirmatrelvir 1, showing P1-P4 moieties and structures of compounds 2-4.
SARS-CoV-2 is an enveloped single-stranded RNA virus whose genome sequencing revealed nearly 82% genome similarities to previous SARS-CoV-1.20,21 Also, SARS-CoV-2 shows 90% resemblance to SARS-CoV-1 in essential enzymes, including Mpro or chymotrypsin-like protease (3CLpro).22,23 Both Mpro and PLpro are cysteine protease, and they process the CoV replicase polyproteins PP1a and PP1b into non-structural proteins essential for viral replication and transcription of the genome. The homodimeric enzyme main protease is highly conserved. It contains a Cys-His catalytic dyad in its active site where the nucleophilic thiolate Cys145 has been exploited in inhibitor design by forming a covalent bond with a range of electrophilic functionalities.24,25 The past and present research efforts focus on the development of covalent inhibitors with electrophilic warhead groups such as: α-ketoamides, aldehydes, esters, α-hydroxymethylketones and benzothiazoles.26,27 Nirmatrelvir is the first orally active main protease inhibitor drug which is a peptidomimetic derivative with a nitrile functionality as the warhead.14,15 The other key features of nirmatrelvir include, a trifluoro acetamide group, two peptide bonds, and a gem-dimethyl cyclopropyl proline derivative which is inherent to Hepatitis C virus protease inhibitor, boceprevir. Incidentally, nirmatrelvir has been designed and developed based upon X-ray structural studies of boceprevir-bound main protease and other structural leads.28,29 Nirmatrelvir was reported to inhibit SARS-CoV-2 Mpro with an inhibitory Ki value of 3.1 nM and antiviral EC50 of 74.5 nM in VeroE6 cell lines when co-treated with the P-glycoprotein inhibitor CP-100356 to prevent drug efflux.14,15 Nirmatrelvir is clinically used as a combination therapy (named Paxlovid) with ritonavir, a well-known pharmacokinetic booster which inhibits CYP3A4 enzyme to slow down metabolic degradation.30,31 Paxlovid has become the drug of choice for treatment of SARS-CoV-2 infection and COVID-19, but it is far from ideal as it presents significant risks of drug -drug interactions and viral rebound-effect.32,33 Therefore, it is very important to develop better antiviral drugs with improved drug properties. As part of our continuing studies for the development of potent and effective coronavirus protease inhibitor, we have investigated the effect of various P4-substituents of nirmatrelvir in combination with 5-membered and 6-membered P1 lactam ring to improve Mpro inhibitory and antiviral activity of the corresponding derivatives. Herein, we report the results of studies which show that several derivatives with a larger 6-membered P1 lactam ring exhibited very potent SARS-CoV-2 Mpro inhibitory activity as well as potent low nanomolar antiviral activity. To obtain insight into the ligand-binding site interactions, we have determined high resolution X-ray crystal structure of the inhibitor-bound to the main protease of SARS-CoV-2.
2. Results and discussion
We and others have determined high resolution X-ray structure crystal of SARS-CoV-2 Mpro and nirmatrelvir complex.14,34 The key interactions between nirmatrelvir and the Mpro active site are highlighted in Figure 2. The warhead P1’ nitrile functionality forms a covalent bond with the active site Cys145, resulting in the formation of a thioimidate adduct in the S1 and S1’ subsites. The P1 5-membered lactam NH forms a hydrogen bond with Glu166 side chain carboxylic acid. The lactam carbonyl oxygen also forms a hydrogen bond with the His163 located in the S1 subsite. The P2 bicycloproline and P3 tert-leucine form a dipeptide bond in nirmatrelvir. The bicycloproline nests in the hydrophobic pocket surrounding His41, Met49, Tyr54, Met165, Asp187, Arg188, and Gln189, the P3 tert-butyl group is mostly solvent exposed. The P3 amide carbonyl oxygen forms a hydrogen bond with backbone NH of Glu166 while the P4 trifluoro acetamide carbonyl oxygen forms a hydrogen bond with Glu166 amide in S4 subsite. Thus, the formation of a covalent bond with Cys145 and extensive ligand binding site interactions are responsible for nirmatrelvir inhibitory activity. 14,34 Based upon this structure analysis, we speculated that a six-membered P1-lactam could fill in the hydrophobic pocket in the S1 subsite more effectively while maintaining the key hydrogen bonding interactions with His163 and Glu166. We have further speculated that other non-polar P4 groups may fill in the S4 subsite effectively and improve potency. For these studies, we synthesized and evaluated both 5 and 6 membered lactams in combination with various P4 groups and compared in vitro activity against nirmatrelvir.
Figure 2.
X-ray crystal structure of Nirmatrelvir (orange) bound to SARS-CoV-2 Main protease (grey) (PDB 7RFW).
Our synthetic strategy for the preparation of various P4 and P1 modified derivatives of nirmatrelvir is shown in Scheme 1. We planned to couple carboxylic acid derived from methyl ester 6a-f with amino nitrile derivative derived from lactam derivatives 7 and 8. The synthesis of lactam derivative 7 was achieved using the recent protocol developed by us.35 The synthesis of 6-membered lactam derivative 8 was carried out using commercially available N-Boc-L-(+)-glutamic acid dimethyl ester 9 as shown in Scheme 2. Treatment of diester 9 with 2.2 equivalents of LiHMDS at −78°C provided the corresponding enolate which was alkylated with bromopropionitrile to furnish alkylated product 10 in 40% yield as a single diastereomer.36,37 The high degree of diastereoselectivity and stereochemical outcome was previously reported by Hanessian and co-workers.38 Selective reduction of nitrile group with a sodium borohydride-cobaltous chloride complex provided the corresponding amine which cyclized to 6-membered lactam derivative 11 in 68% yield. Methyl ester 11 was then converted to nitrile derivative 8 in a two-step sequence. Reaction of 11 with ammonium hydroxide in MeOH at 23 °C for 12 h provided the corresponding carboxamide derivative. The resulting carboxamide was treated with tosyl chloride and pyridine at 23°C for 18 h to furnish the nitrile derivative 8 in 58% yield over 2-steps.
Scheme 1.
Synthetic strategy for inhibitors 5a-i
Scheme 2.
Asymmetric synthesis of nitrile derivative 8.
The synthesis of P4 carboxamide derivatives in combination with the P1 5- and 6-membered lactam rings are achieved with minimum racemization of the tert-leucine side chain utilizing the synthetic route developed by us recently for the synthesis of nirmatrelvir.35 As shown in Scheme 3, dipeptide methyl ester 6a was prepared in multigram quantity by coupling commercially available optically active bicyclic proline methyl ester 12 with Boc-tert-leucine 13a as described previously.15 Treatment of Boc-derivative 6a with 10% ZnCl2 and acetic anhydride in CH2Cl2 at 23 °C for 12 h provided the acetamide derivative 14a in 81% yield with minimum racemization (< 5%). Similarly, reaction of 6a with ZnCl2 and benzoic anhydride, pivalic anhydride, trifluoroacetic anhydride, and trichloroacetic anhydride furnished the respective amide derivatives 14b-e in good to excellent yields (74–96%). Saponification of these methyl esters with LiOH in THF/H2O at 23 °C for 1 h yielded (61–95%) the corresponding acids 15a-e. The amine coupling partners were prepared from the corresponding aminonitrile derivatives 7 and 8 via Boc-deprotection using trifluoroacetic acid (TFA). The resulting amines were coupled with acids 15a-e using HATU and NMM in DMF at 23°C for 12 h furnished inhibitors 5a-f in moderate to good yields (21–64%).
Scheme 3.
Synthesis of inhibitors 5a-f.
Synthesis of various P4-carbamate derivatives and bis-trifluoroacetamide derivatives is shown in Scheme 4. Coupling of bicycloproline derivative 12 with P4-substituted tert-leucine derivatives 13a-f provided the carbamate derivatives 6a-f in good yields. Saponification of 6a with aqueous LiOH at 23 °C for 1 h followed by coupling of the resulting acid with amine derived from nitrile 7 furnished Boc-derivative 5g in 62% yield. We attempted to convert Boc-derivative 5g to nirmatrelvir using our previously described procedure with ZnCl2 and trifluoroacetic anhydride.35 Interestingly, reaction of 5g with 10% ZnCl2 in the presence of trifluoracetic anhydride at 23 °C for 12 h did not provide nirmatrelvir 1, instead bis-amide derivative 5h was obtained in 85% yield. Carbamate derivatives 6b-f were converted to various P1 5-membered lactam analogs 5i-m using a two-steps sequence involving saponification of esters 6b-f followed by coupling of the resulting acids with amine derivative from 7 after removal of Boc-group with trifluoroacetic acid. Carbamate derivatives 5g-m were obtained in good yields. Similarly, 3-pyridyl and 4-pyridyl carbamates 6d and 6e were converted to compounds 5n and 5o, respectively by saponification of 6d and 6e followed by coupling of the resulting acids with amine derived from 8 after treatment with TFA as described above. Both carbamates 5n and 5o were obtained in 34% yield.
Scheme 4.
Synthesis of inhibitors 5g-o.
We have prepared a series of P4-N-amide derivatives to assess stereoelectronic effect and hydrogen bonding capabilities of the amide NH group in the S4 subsite. The structure and biological activity of these inhibitors are shown in Table 1. We utilized our previously reported assay protocols for SARS-CoV-2 Mpro inhibitory activity and antiviral activity in veroE6 cells.39, 40 As shown in Table 1, in-house prepared nirmatrelvir 1, exhibited a SARS-CoV-2 Mpro inhibitory activity of 0.26 nM. It showed an antiviral EC50 value of 2.0 μM. Remdesivir displayed antiviral EC50 of 3.2 μM in the same assay. In comparison nirmatrelvir was reported to have an average Ki value of 3.1 nM and antiviral activity of 74.5 nM in veroE6 cells.14 We examined the effect N-acetamide group in compound 5a in place of the N-trifluoroacetamide group in 1. Compound 5a exhibited comparable Mpro inhibitory activity to 1 (entry 2). The incorporation of N-benzamide group, 5b, also resulted in substantial loss of both Mpro inhibitory and antiviral activity (entry 3). Replacement of the trifluoroacetamide with the bulky pivalamide group in compound 5c caused nearly 10-fold loss of Mpro inhibitory activity. However, compound 5c exhibited comparable antiviral activity, EC50 2.3 μM vs 2.0 μM for 1 (entries 1 and 4). We next examined the effect of increasing the size of the halogen from N-trifluoro to N-trichloro group in compound 5d. This replacement resulted in significant improvement of both Mpro inhibitory and antiviral activity. Compound 5d displayed very similar in vitro activity as nirmatrelvir 1 (entries 1 and 5). We then investigated the effect of a 6-membered lactam at P1 position as it evident from the X-ray crystal structure of SARS-CoV-2 Mpro bond nirmatrelvir that a larger ring can be accommodated in the S1-subsite.
Table 1.
SARS-CoV-2 3CLpro inhibition by nirmatrelvir derivatives 5a-h.
| Entry | Inhibitor | Ki (nM)a | EC50 (μM)b |
|---|---|---|---|
| 1 |
|
0.26 | 2.0 |
| 2. |
|
0.44 | 4.0 |
| 3 |
|
36.9 | 17 |
| 4 |
|
22.1 | 2.3 |
| 5 |
|
0.2 | 1.6 |
| 6 |
|
0.04 | 0.26 |
| 7 |
|
1.1 | 0.27 |
| 8. |
|
32,300 | 100 |
Ki values were determined from fitting inhibition data to the Morrison equation as described.39
EC50 values are means of at least four experiments.
Indeed, compound 5e containing a P4 6-membered lactam led to more than 6-fold improvement of Mpro inhibitory activity as well as 7-fold improvement in antiviral activity over nirmatrelvir 1 (entry 6). Incorporation of P4 trichloromethyl group in place of trifluoromethyl provided inhibitor 5f which displayed very potent Mpro inhibitory activity (Ki 1.1 nM) and over 7-fold improvement in antiviral activity over nirmatrelvir 1 (entry 7). We have also evaluated P2 trifluoroacetamide derivative 5h. However, this compound showed no appreciable Mpro inhibitory or antiviral activity. This result suggests the important role of the P2 NH group for binding of nirmatrelvir in the Mpro active site.
We have also investigated the feasibility of various P4 carbamate and urea derivatives that can impact steric and electronic effects. The X-ray crystal structure of nirmatrelvir-bound to SARS-CoV-2 Mpro suggests that the P4 carbamate NH group can effectively form a hydrogen bond with the Glu166 backbone carbonyl group. Also, a suitable alkyl or aryl group could fill the S4 subsite. The biological evaluation results are shown in Table 2. The P4 tert-butyloxycarbonyl group containing inhibitor 5g showed a significant loss in Mpro inhibitory and antiviral activity as compared to nirmatrelvir 1 (entry 1). Incorporation of a benzyloxy carbonyl group in compound 5i resulted in significant improvement of Mpro inhibitory activity compared to the Boc-group containing derivative 5g (entry 2). Compound 5i also exhibited very potent antiviral activity with EC50 value of 290 nM. The effect of pyridyl methyl group over the benzyl group was then evaluated in compounds 5j-l. Interestingly, the 3-pyridyl methyl carbamate 5k displayed significantly enhanced activity over the 4-pyridylmethyl carbamate 5j and 2-pyridylmethyl carabamate 5l (entries 3–5). Carbamate 5k also exhibited very potent antiviral activity with an EC50 value of 35 nM, which is over an 8-fold improvement of antiviral activity compared to benzyl carbamate derivative 5i. The current structure-activity relationship (SAR) indicates that the benzyloxy group fills the S4 subsite more effectively than the tert-butyloxy group in 5g. Also, the position of nitrogen atom in the pyridyl ring is very important. However, the actual contribution of the 3-pyridyl group in compound 5k in the Mpro active site is not clear. Further expansion of SAR and X-ray structural studies are the subject of current investigation. Incorporation of tert-butyl urea at P4 resulted in compound 5m which exhibited potent Mpro inhibitory activity. However, this compound exhibited 3-fold loss of antiviral activity compared to the corresponding Boc-derivative 5g (entries 1 and 6). We have examined the effect of P4 3-and 4-pyridyl carbamate in combination with 6-membered lactam. Compounds 5n with 3-pyridylmethyl carbamate showed comparable Mpro inhibitiory activity over compound 5k with 5-membered lactam. However, compound 5n displayed significant loss of antiviral activity over 5k (entries 4 and 7). Carbamate derivative 5o containing 4-pyridylmethyl carbamate showed 4-fold improvement of Mpro inhibitory activity but exhibited comparable antiviral activity (entries 3 and 8). Overall, benzyl and 3-pyridylmethyl carbamates 5i, 5k, and amide derivatives 5e, 5f exhibited very potent antiviral activity comparable to nirmatrelvir. We have also examined and compared anti-SARS-CoV-2 activity of compound 5f against variants of concern and the result of this investigation is shown in Table 3.41 Interestingly, compound 5f maintained excellent antiviral activity against Alpha and Delta SARS-CoV-2 variants. However, compound 5f lost 3–5-fold antiviral activity against beta, gamma and omicron variants. Remdesivir and nirmatrelvir displayed comparable antiviral activity.
Table 2.
SARS-CoV-2 3CLpro inhibition by nirmatrelvir derivatives 5g-o.
| Entry | Inhibitor | Ki (nM)a | EC50 (μM)b |
|---|---|---|---|
| 1 |
|
7.7 | 30.7 |
| 2 |
|
0.025 | 0.29 |
| 3. |
|
32.2 | 29.7 |
| 4. |
|
0.09 | 0.035 |
| 5. |
|
0.49 | 40.1 |
| 6. |
|
3.3 | 100 |
| 7. |
|
0.094 | 3.4 |
| 8. |
|
7.8 | 27.1 |
Ki values were determined from fitting inhibition data to the Morrison equation as described.39
EC50 values are means of at least four experiments.
Table 3.
Anti-SARS-CoV-2 activity of compound 5f against VOCs (variants of concern)
| EC50(μM) | ||||||
|---|---|---|---|---|---|---|
| Compound | SARS-CoV-205−2N (Wuhan) | SARS-CoV-2QK2 (Alpha) | SARS-CoV-2TY8 (Beta) | SARS-CoV-2TY7 (Gamma) | SARS-CoV-2K1734 (Delta) | SARS-CoV-2929 (Omicron) |
| Compound 5f (GRL-018–22) | 0.7 ± 0.1 | 0.8 ± 0.2 | 2.40 ± 0.09 | 2.0 ± 0.1 | 0.6 ± 0.2 | 4.0 ± 1.2 |
| Remdesivir | 3.8 ± 1.9 | 3.5 ± 0.1 | 2.8 ± 0.1 | 2.6 ± 0.1 | 2.9 ± 0.1 | 4.2 ± 0.5 |
| Nirmatrelvir | 3.0 ± 0.1 | 2.5 ± 0.1 | 3.7 ± 0.2 | 4.0 ± 0.7 | 2.9 ± 0.2 | 3.1 ± 0.2 |
EC50 values were determined by WST-8 assays, using VeroE6 cells. Data was collected on day-3 post infection (day-7, Omicron).
We also evaluated compound 5e in our immunocytochemistry assay to assess antiviral activity and comapre with the activity of nirmatrelvir at the cellular level. We utilized primary antibody IgG fraction isolated from a COVID-19 convalescent patient in this assay. These patients possess high titers of neutralizing antibodies as well as SARS-CoV-2-binding IgG antibodies.40,42 As can be seen in Figure 3, robust cellular cytoskeleton filamentors actin (F-actin) were observed as mesh-like structures (in red) and a lot of nuclei (in blue) were formed which suggest that cells were healthy and replicating. As shown, the exposure of VeroE6 cells to SARS-CoV-2WK−521 resulted in the destruction of the F-actin structure and viral infection of the cells (stained green, top right panel). However, when VeroE6 cells are exposed to SARS-CoV-2WK−521 in the presence of 100 μM and 10 μM concentration of compound 5e and nirmatrelvir, there were hardly any infected cells. While some infected cells were observed in the presence of 1 μM compound 5e, nirmatrelvir exposed culture at 1 μM showed complete viral breakthrough (infected cells, stained green). Compound 5e exerted potent Mpro inhibitory activity (Ki 40 pM) and antiviral activity (EC50 0.26 μM). Also, it significantly reduced the infectivity, replication, and cytopathic effect of SARS-CoV-2 without significant toxicity.
Figure 3.

Inhibitor 5e and nirmatrelvir potently blocked the infectivity and cytopathic effect of SARS-CoV-2WK−521 in VeroE6TMPRSS2 cells.
To better understand the molecular interactions of the 6-membered lactam of inhibitor 5e with the SARS-CoV-2 Mpro active site, we determined the X-ray structure of Mpro in complex with 5e to 1.9 Å resolution.43 Our X-ray crystallography data clearly shows strong and continuous electron density of compound 5e inside the Mpro active site as shown in Figure 4. The overall structure is very similar to that of the nirmatrelvir SARS-CoV-2 Mpro complex.14 The key interactions between inhibitor 5e and Mpro active site residues are highlighted in Figure 5. Compound 5e occupies the substrate binding pockets from S1 to S4. It forms seven direct hydrogen bonds with various Mpro residues. In particular, compound 5e forms two hydrogen bonds with the backbone carbonyl and amide NH of Glu166 involving the P4 amide NH and P3 amide carbonyl group respectively. The backbone carbonyl of His164 also forms a hydrogen bond with the P2 amide NH of inhibitor 5e. The six membered lactam NH forms two hydrogen bonds with Glu166 side chain carboxylate as well as the backbone carbonyl of Phe140. The lactam carbonyl group accepts a hydrogen bond from the His163 side chain imidazole ε2-nitrogen assuming it is protonated. The active site Cys145 sulfur atom performs a nucleophilic attack on the nitrile functionality of 5e forming a covalent bond, and the resulting imidate forms a hydrogen bonds with the backbone NH of Cys145. Apart from hydrogen bonding interactions, compound 5e fills the hydrophobic pockets particularly the S1-S3 subsites of Mpro active site and is involved in van der Waals interactions.
Figure 4.
X-ray crystal structure of SARS-CoV-2 3CLpro covalently bound to compound 5e through Cys145 (PDB:8UND). (Left Panel) Polder electron density omit map (mFobs − DFmodel ) surrounding 5e contoured at (+) 4.0 σ and represented as a solid surface colored in Khaki. The enzyme 3CLpro is depicted as a gray surface.
Figure 5.
X-ray structure of inhibitor 5e-bound to SARS-CoV-2 Mpro. The inhibitor carbon atoms are shown in orange and the Mpro carbon atoms are shown in grey. Hydrogen bond interactions between 5e and Mpro are indicated by dotted lines (PDB ID:8UND).
The main difference between nirmatrelvir and inhibitor 5e is the presence of a six-membered lactam in place of the 5-membered lactam in 1. We therefore compared both hydrogen bonding distances and van der Waals interactions of the 5-membered lactam of nirmatrelvir and the six-membered lactam of inhibitor 5e in the Mpro active site. This structural analysis is shown in Figure 6. Interestingly, the P1 lactam carbonyl and NH groups of inhibitors 5e form very strong hydrogen bonds with the His163 side chain, Glu166 carboxylic acid chain, and Phe140 backbone NH, (2.7 Å, 2.7 Å and 3.2 Å respectively, Figure 5A) compared to 5-membered lactam in 1 (2.8 Å. 3.2 Å and 3.2 Å respectively, Figure 5B). The lactam rings in both compounds 1 and 5e engage in van der Waals interaction with Asn142, Phe140 and Gly141. The extra methylene group in the lactam ring of 5e makes stronger van der Waals interaction with these residues in the S1 subsite. Both enhanced hydrogen bonding and van der Waals interactions of six membered lactam in compound 5e contributed to the high Mpro affinity and antiviral activity compared to inhibitor 1.
Figure 6.
Comparison of the P1 six-membered lactam moiety of inhibitor 5e (5A-top, orange carbon atoms) with the P1 5-membered lactam moiety of nirmatrelvir (5B-bottom, cyan carbon chain) inside the S1 subsite. Both ligands form extensive van der Waals interactions in the S1 subsite. Also, both inhibitors form strong hydrogen bonds in a similar fashion (black dotted lines).
3. Conclusions
In summary, we have modified P1 and P4 ligands of nirmatrelvir, an FDA approved drug for COVID-19 treatment. Our investigation has been aimed at improving inhibitor activity by modification of inhbitor interactions within the S1 subsite and filling in the hydrophobic pocket in the S4 subsite. In particular, we examined various alkyl, aryl, halogenated acetamide, carbamate and urea derivatives as the P4 ligands in combination with 5-membered and 6-membered lactams as the P1 ligands. The P4 amide derivatives are significantly more potent than carbamate and urea derivatives examined in this study. Several inhibitors with a larger 6-membered lactam ring with halogenated P4 amides showed significant enhancement of inhibitory and antiviral activity over nirmatrelvir. Compound 5e with a P1 6-membered lactam and P4 trifluoro acetamide groups exhibited 3-fold improvement in antiviral activity in VeroE6 cells. This compound maintained very good activity against variants concerns such as Delta and Omicron variants, maintaining similar antiviral activity as remdesivir. To obtain insight into the ligand-binding site interactions for the 6-membered lactam in the S1 subsite, we determined the high resolution X-ray crystal structure of inhibitor 5e-bound to SARS-CoV-2 Mpro. The structural analysis showed that the 6-membered lactam ring functionality forms stronger hydrogen bonding compared to the 5-membered lactam in nirmatrelvir in the S1 subsite. The 6-membered lactam ring also fills the hydrophobic pocket more effectively than nirmatrelvir’s 5-membered lactam ring. Our investigation revealed the potency enhancing effect of the P1 6-membered lactam in this series of compounds. The current result will further aid the optimization of nirmatrelvir derivatives and related compounds for Mpro inhibition. Design and evaluation of novel Mpro inhibitors are in progress in our laboratories.
4. Experimental Section
All chemicals and solvents were purchased from commercial suppliers and were used as received unless otherwise stated. All reactions were carried out under an argon atmosphere in either flame or oven-dried (120 °C) glassware. TLC analysis was conducted using glass-backed thin-layer silica gel chromatography plates (60 Å, 250 μm thickness, F254 indicator). Column chromatography was performed using silica gel, 230– 400 mesh, 60 Å pore diameter. Isolated yields and yields based on the recovered starting material (brsm) were determined following purification. Proton Nuclear Magnetic Resonance NMR (1H NMR) spectra and carbon nuclear magnetic resonance (13C NMR) spectra were recorded on Bruker AV-III-400HD and Bruker AVIII-800 spectrometers. Chemical shifts for protons are reported in parts per million and are references to the NMR solvent peak (CDCl3: δ 7.26). Chemical shifts for carbons are reported in parts per million and are referenced to the carbon resonances of the NMR solvent (CDCl3: δ77.16). Data are reported as (s = singlet, d = doublet, t = triplet, q = quartet, quint = quintet, sep = septet, m = multiplet, dd = doublet of doublets, ddd = doublet of doublet of doublets, dddd = doublet of doublet of doublets of doublets, td = triplet of doublets, dq = doublet of quartets, qd = quartet of doublets, dt = doublet of triplets, brs = broad singlet). All coupling constants are measured in hertz (Hz). Optical rotations were measured on a Rudolph’s AUTOPOL-III automatic digital polarimeter with a sodium lamp and are reported as follows: [α]λ T °C (c = g/100 mL, solvent). High-resolution mass spectrometry (HRMS) spectra were recorded under positive electron spray ionization (ESI+) at Agilent 6550 Q-TOF LC/MS instrument at the Purdue University Analytical Mass Spectrometry Facility.
Dimethyl (2S,4S)-2-((tert-butoxycarbonyl)amino)-4-(2-cyanoethyl)pentanedioate (10):
To a solution of N-Boc-L(+)-gluatamic dimethyl ester 9 (3.0 g, 10.9 mmol) in anhydrous THF (30 mL) was added the solution of lithium hexamethyldisilazide (1M solution in THF, 24 mL, 23.56 mmol) dropwise under argon atmosphere at −78 °C. After 1 h of stirring at −78 °C, 3-bromopropionitrile (2.19 g, 1.36 mmol) solution in anhydrous THF (10 mL) was added dropwise to the reaction mixture while maintaining the temperature under −78 °C. The reaction mixture was stirred at −78 °C for an additional 2 h and quenched with cold methanol (30 mL). The crude mixture was stirred for 30 minutes at −78 °C followed by addition of cold solution of 1:6 mixture of acetic acid and THF (3 mL AcOH/ 18 mL THF). After a further 30 min of stirring at −78 °C, the cooling bath was removed, and the reaction mixture was allowed to warm up to 23 °C. The crude mixture was diluted with EtOAc, washed with brine. The organic phase was dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (25% EtOAc/hexanes) to give the ester 10 (1.42 g, 40%) as clear oil. 1H NMR (400 MHz, CDCl3) δ 5.03 (d, J = 8.7 Hz, 1H), 4.44 – 4.27 (m, 1H), 3.73 (s, 3H), 3.70 (s, 3H), 2.67 – 2.55 (m, 1H), 2.38 (td, J = 7.2, 4.1 Hz, 2H), 2.07 – 1.90 (m, 4H), 1.43 (s, 9H); ESI-MS (m/z): [M + H]+ : 329.1.
Methyl (S)-2-((tert-butoxycarbonyl)amino)-3-((S)-2-oxopiperidin-3-yl)propanoate (11):
To a vigorously stirring solution of ester 10 (1.30 g, 3.96 mmol) in anhydrous MeOH (26 mL) was added CoCl2·6H2O (565 mg, 2.38 mmol). The resulting pink solution was cooled to 0 °C and NaBH4 (903 mg, 23.76 mmol) was added portion-wise. After 15 minutes, the ice bath was removed, and the reaction mixture was warmed to 23 °C. After 20 h, the solvents were evaporated in vacuo and the crude mixture was poured into a cold solution of 1M citric acid (50 mL). The residual mixture was extracted with ethyl acetate (3 × 20 mL) and the combined organic layers were washed with saturated sodium bicarbonate solution and brine. The organic phase was dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (50–100% EtOAc/hexanes) to give the lactam 11 (800 mg, 68%) as fluffy white solid. 1H NMR (400 MHz, CDCl3) δ 6.19 (s, 1H), 5.62 (d, J = 8.5 Hz, 1H), 4.46 – 4.19 (m, 1H), 3.71 (s, 3H), 3.29 (dp, J = 6.9, 2.2 Hz, 2H), 2.38 – 2.22 (m, 2H), 2.13 (dd, J = 11.2, 5.5 Hz, 1H), 1.87 (s, 1H), 1.84 – 1.69 (m, 2H), 1.60 – 1.50 (m, 1H), 1.42 (s, 9H); ESI-MS (m/z): [M + H]+ : 301.1.
Tert-butyl ((S)-1-cyano-2-((S)-2-oxopiperidin-3-yl)ethyl)carbamate (8):
Boc-protected amino ester 11 (300 mg, 1.0 mmol) was dissolved in a 1: 1 mixture of MeOH (5.0 mL) and NH4OH (28–30% NH3, 5.0 mL) at 23 °C. The reaction mixture was stirred for 12 h. Upon completion, the solvents were evaporated in vacuo and the crude primary amide was used for the subsequent reaction without further purification. To a solution of the above primary amide (285 mg, 1.0 mmol) in 2.5 mL CH2Cl2 at 0 °C were added p-toluene sulfonyl chloride (381 mg, 2.0 mmol) and pyridine (402 μL, 5.0 mmol). The resulting solution was warmed to 23 °C and stirred for 18 h. The reaction was quenched by saturated sodium bicarbonate solution and extracted with CH2Cl2 (3 × 5 mL). The combined organic layers were washed with 1N HCl, saturated sodium bicarbonate solution and brine. The organic phase was dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (0–2% MeOH/CH2Cl2) to give the nitrile 8 (150 mg, 58% over two steps) as fluffy white solid. 1H NMR (400 MHz, CDCl3) δ 6.61 (s, 1H), 6.21 (d, J = 7.6 Hz, 1H), 4.68 (d, J = 8.6 Hz, 1H), 3.29 (ddt, J = 9.0, 4.8, 2.1 Hz, 2H), 2.46 – 2.26 (m, 2H), 2.10 – 1.96 (m, 1H), 1.87 (dq, J = 13.6, 4.5 Hz, 2H), 1.80 – 1.64 (m, 1H), 1.55 (ddd, J = 13.8, 8.1, 3.0 Hz, 1H), 1.43 (s, 9H); 13C NMR (100 MHz, CDCl3) δ 173.9, 154.8, 119.1, 80.7, 42.1, 40.9, 37.7, 34.9, 28.2, 27.1, 21.5; HRMS (ESI/LTQ) m/z: [M + H]+ calcd for C13H22N3O3 268.16609, found: 268.16614.
Methyl(1R,2S,5S)-3-((S)-2-((tert-butoxycarbonyl)amino)-3,3-dimethylbutanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate (6a):
To a stirred solution of Boc-L-tert-leucine 13a (1.35 g, 5.85 mmol) and methyl (1R,2S,5S)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate hydrochloride 12 (1.0 g, 4.88 mmol) in 40 mL DMF at 0 °C were added EDC·HCl (1.12 g, 5.85 mmol), HOBt (896 mg, 5.85 mmol), and N-methylmorpholine (1.30 mL, 12.20 mmol). After 10 minutes, the ice bath was removed, and the reaction mixture was stirred at 23 °C for 5 h. The reaction then diluted with diethyl ether and washed with cold H2O. The combined organic layers were washed with brine, dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (5–10% EtOAc/hexanes) to give the dipeptide methyl ester 6a (1.37 g, 74%) as white sticky solid. [α]D20 −88.0 (c 1.73, CHCl3). 1H NMR (400 MHz, DMSO-d6) δ 8.29 (s, 1H), 6.69 (d, J = 8.0 Hz, 1H), 4.19 (s, 1H), 4.04 (d, J = 12.0 Hz, 1H), 3.92 (d, J = 12.0 Hz, 1H), 3.79 – 3.63 (m, 1H), 3.29 (s, 3H), 1.52 – 1.48 (m, 1H), 1.40 – 1.38 (m, 1H), 1.33 (s, 9H), 0.99 (s, 3H), 0.92 (s, 9H), 0.83 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 171.9, 170.5, 156.3, 78.6, 59.1, 58.9, 52,3, 47.4, 34.4, 29.9, 28.4, 27.4, 26.6, 26.1, 19.3, 12.5; HRMS (ESI/LTQ) m/z: [M + H]+ calcd for C20H35N2O5 383.2546, found: 383.2548.
Methyl(1R,2S,5S)-3-((S)-2-(((benzyloxy)carbonyl)amino)-3,3-dimethylbutanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate (6b):
To a stirred solution of Cbz-L-tert-leucine 13b (100 mg, 0.377 mmol) and methyl (1R,2S,5S)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate hydrochloride 12 (78 mg, 0.377 mmol) in 3.0 mL of DMF at 0 °C were added HATU (172 mg, 0.452 mmol) and DIPEA (0.076 mL, 0.452 mmol). After 10 minutes, the ice bath was removed, and the reaction mixture was stirred at 23 °C for 12 h. The reaction then diluted with diethyl ether and washed with cold H2O. The combined organic layers were washed with brine, dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (1–2% MeOH/CH2Cl2) to give the carbamate derivative 6b (112 mg, 71%) as white fluffy solid. 1H NMR (400 MHz, CDCl3) δ 7.34 – 7.29 (m, 5H), 5.37 (d, J = 10.0 Hz, 1H), 5.06 (d, J = 5.4 Hz, 2H), 4.45 (s, 1H), 4.26 (d, J = 10.0 Hz, 1H), 3.90 (t, J = 2.8 Hz, 2H), 3.74 (s, 3H), 1.44 (s, 1H), 1.02 (s, 12H), 0.94 (s, 1H), 0.86 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.8, 170.4, 156.3, 136.3, 128.4, 127.8, 66.8, 59.2, 59.1, 52.2, 47.6, 35.2, 30.2, 27.3, 26.2, 19.3, 12.4; LRMS-ESI (m/z) [M+H]+ 417.5.
Methyl (1R,2S,5S)-3-((S)-3,3-dimethyl-2-(((pyridin-2-ylmethoxy)carbonyl)amino)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate (6c):
Following the procedure described for the preparation of carbamate derivative 6b using (S)-3,3-dimethyl-2-(((pyridin-2-ylmethoxy)carbonyl)amino)butanoic acid 13c (181 mg, 0.68 mmol) and proline derivative 12 (140 mg, 0.68 mmol)afforded the acid 6c (200 mg, 71%) as a brown oil. 1H NMR (400 MHz, CDCl3) δ 8.57 – 8.56 (m, 1H), 7.67 (td, J = 7.7, 1.8 Hz, 1H), 7.29 (d, J = 7.8 Hz, 1H), 7.20 (dd, J = 7.5, 5.1 Hz, 1H), 5.48 (d, J = 10.1 Hz, 1H), 5.18 (d, J = 4.1 Hz, 2H), 4.46 (s, 1H), 4.27 (d, J = 10.0 Hz, 1H), 3.90 – 3.89 (m, 2H), 3.75 (s, 3H), 1.45 (s, 1H), 1.04 (s, 9H), 1.03 (s, 3H), 0.96 (s, 1H), 0.87 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.8, 170.3, 156.3, 156.0, 149.2, 136.6, 122.6, 121.2, 67.3, 59.2, 52.2, 47.6, 35.3, 30.2, 27.3, 26.2, 19.4, 12.4; LRMS-ESI (m/z) [M+H]+ 418.2.
Methyl (1R,2S,5S)-3-((S)-3,3-dimethyl-2-(((pyridin-3-ylmethoxy)carbonyl)amino)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate (6d):
Following the procedure described for the preparation of carbamate derivative 6b using (S)-3,3-dimethyl-3-(((pyridin-2-ylmethoxy)carbonyl)amino)butanoic acid 13d (194 mg, 0.729 mmol) and proline derivative 12 (150 mg, 0.729 mmol) afforded the acid 6d (220 mg, 78%) as a brown oil. 1H NMR (400 MHz, CDCl3) δ 8.59 (s, 2H), 7.66 (dd, J = 7.8, 1.9 Hz, 1H), 7.29 – 7.27 (m, 1H), 5.39 (d, J = 9.9 Hz, 1H), 5.08 (d, J = 4.8 Hz, 2H), 4.45 (s, 1H), 4.24 (d, J = 10.0 Hz, 1H), 3.89 (d, J = 2.8 Hz, 2H), 3.74 (s, 3H), 1.45 (s, 1H), 1.03 (s, 3H), 1.03 (s, 9H), 0.94 (s, 1H), 0.85 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.7, 170.3, 156.0, 149.4, 135.6, 123.3, 64.2, 59.2, 52.2, 47.6, 35.2, 30.2, 27.3, 26.2, 19.3, 12.3; LRMS-ESI (m/z) [M+H]+ 418.2.
Methyl(1R,2S,5S)-3-((S)-3,3-dimethyl-2-(((pyridin-4-ylmethoxy)carbonyl)amino)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate (6e):
Following the procedure described for the preparation of carbamate derivative 6b using (S)-3,3-dimethyl-4-(((pyridin-2-ylmethoxy)carbonyl)amino)butanoic acid 13e (65 mg, 0.243 mmol) and proline derivative 12 (50 mg, 0.243 mmol) afforded the acid 6e (72 mg, 71%) as a brown oil. 1H NMR (400 MHz, CDCl3) δ 8.57 – 8.55 (m, 2H), 7.20 – 7.18 (m, 2H), 5.48 (d, J = 10.0 Hz, 1H), 5.13 – 5.01 (m, 2H), 4.46 (s, 1H), 4.24 (d, J = 10.0 Hz, 1H), 3.88 – 3.87 (m, 2H), 3.75 (s, 3H), 1.44 (d, J = 1.7 Hz, 1H), 1.04 (s, 9H), 1.02 (s, 3H), 0.95 (s, 1H), 0.83 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.7, 170.2, 155.8, 149.9, 145.5, 121.4, 64.8, 59.3, 59.2, 52.2, 47.7, 35.3, 30.2, 27.3, 26.2, 26.1, 19.3, 12.3; LRMS-ESI (m/z) [M+H]+ 418.2.
Methyl(1R,2S,5S)-3-((S)-2-acetamido-3,3-dimethylbutanoyl)-6,6-dimethyl-3-azabicyclo [3.1.0]hexane-2-carboxylate (14a):
To a stirred solution of methyl (1R,2S,5S)-3-((S)-2-((tert-butoxycarbonyl)amino)-3,3-dimethylbutanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate 6a (100 mg, 0.261 mmol) in 3 mL CH2Cl2 at 23 °C were added ZnCl2 (13 mg, 0.10 mmol) and acetic anhydride (0.1 mL, 1.04 mmol). After 12 h, the reaction mixture was quenched with H2O and extracted with CH2Cl2 (3 × 10 mL). The combined organic layers were washed with brine, dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (30% EtOAc/hexanes) to give acetamide 14a (68 mg, 81%) white sticky solid. 1H NMR (400 MHz, CDCl3) δ 6.01 (d, J = 9.7 Hz, 1H), 4.57 (d, J = 9.6 Hz, 1H), 4.45 (s, 1H), 3.92 – 3.90 (m, 2H), 3.75 (s, 3H), 1.98 (s, 3H), 1.46 (dd, J = 4.0, 1.4 Hz, 1H), 1.04 (s, 3H), 1.03 (s, 9H), 0.94 (s, 1H), 0.89 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.8, 170.3, 169.6, 59.2, 56.9, 52.2, 47.7, 35.3, 30.2, 27.3, 26.2, 26.1, 23.1, 19.4, 12.4; LRMS-ESI (m/z) [M+H]+ 325.2.
Methyl(1R,2S,5S)-3-((S)-2-benzamido-3,3-dimethylbutanoyl)-6,6-dimethyl-3-azabicyclo [3.1.0]hexane-2-carboxylate (14b):
Following the procedure described for the preparation of compound 14a with a slight modification of conducting the reaction with benzoic anhydride (118 mg, 0.52 mmol) and ZnCl2 (18 mg, 0.131 mmol) at 40 °C afforded the benzamide 14b (93 mg, 92%) as a white sticky solid. 1H NMR (400 MHz, CDCl3) δ 7.72 (dt, J = 7.1, 1.4 Hz, 2H), 7.47 – 7.43 (m, 1H), 7.38 (dd, J = 8.2, 6.6 Hz, 2H), 6.70 (d, J = 9.5 Hz, 1H), 4.78 (d, J = 9.6 Hz, 1H), 4.43 (s, 1H), 4.01 – 3.90 (m, 2H), 3.73 (s, 3H), 1.45 (d, J = 4.7 Hz, 1H), 1.08 (s, 9H), 1.00 (s, 3H), 0.99 (s, 1H), 0.84 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.7, 170.2, 166.9, 134.0, 131.5, 128.5, 128.4, 126.9, 59.3, 57.2, 52.2, 47.7, 35.8, 30.2, 27.3, 26.5, 26.4, 26.1, 19.3, 12.3; LS-MS (ESI, m/z): [M + Na]+ : 409.4.
Methyl(1R,2S,5S)-3-((S)-3,3-dimethyl-2-pivalamidobutanoyl)-6,6-dimethyl-3-azabicyclo [3.1.0]hexane-2-carboxylate (14c):
Following the procedure described for the preparation of compound 14a with a slight modification of conducting the reaction with pivalic anhydride (97 mg, 0.52 mmol) and ZnCl2 (18 mg, 0.131 mmol) at 40 °C afforded the tert-butyl amide 14c (71 mg, 74%) as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 6.13 (d, J = 9.6 Hz, 1H), 4.53 (d, J = 9.6 Hz, 1H), 4.40 (d, J = 6.1 Hz, 1H), 3.92 – 3.82 (m, 2H), 3.70 (d, J = 1.7 Hz, 3H), 1.39 (s, 1H), 1.13 (s, 1H), 1.11 (s, 9H), 0.98 (s, 3H), 0.97 (s, 9H), 0.88 (s, 1H), 0.81 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 177.8, 171.8, 170.4, 60.7, 59.1, 56.5, 52.1, 47.5, 38.7, 36.4, 35.3, 32.6, 30.1, 27.3, 26.3, 26.2, 26.1, 19.2, 12.2; LS-MS (ESI, m/z): [M + Na]+ : 389.3.
Methyl (1R,2S,5S)-3-((S)-3,3-dimethyl-2-(2,2,2-trifluoroacetamido)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate (14d):
Following the procedure described for the preparation of compound 14a with trifluoroacetic anhydride (0.76 mL, 5.46 mmol) and ZnCl2 (25 mg, 0.18 mmol) at 23 °C afforded the trifluoroacetamide 14d (623 mg, 91%) as a white sticky solid. [α]D20 −93.75 (c 0.16, CHCl3). 1H NMR (400 MHz, CDCl3) 7.09 (d, J = 9.5 Hz, 1H), 4.52 (d, J = 9.5 Hz, 1H), 4.41 (s, 1H), 3.89 (dd, J = 10.2, 4.6 Hz, 1H), 3.76 (d, J = 10.3 Hz, 1H), 3.70 (s, 3H), 1.43 (s, 1H), 1.01 (s, 9H), 1.00 (s, 3H), 0.96 – 0.93 (m, 1H), 0.83 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.4, 168.3, 156.8 (q, J = 37.6 Hz), 115.7 (q, J = 287.7 Hz), 59.4, 57.6, 52.2, 47.7, 35.9, 30.1, 27.2, 26.2, 26.1, 25.9, 19.3, 12.2; HRMS (ESI/Q-TOF) m/z: [M + H]+ calcd for C17H26F3N2O4 379.1839, found: 379.1840.
Methyl (1R,2S,5S)-3-((S)-3,3-dimethyl-2-(2,2,2-trichloroacetamido)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylate (14e):
Following the procedure described for the preparation of compound 14a with a slight modification of conducting the reaction with trichloroacetic anhydride (153 mg, 0.50 mmol) and ZnCl2 (18 mg, 0.131 mmol) at 40 °C afforded the trichloroacetamide 14e (102 mg, 96%) as a white sticky solid. 1H NMR (400 MHz, CDCl3) δ 7.26 – 7.24 (m, 1H), 4.47 (d, J = 6.9 Hz, 2H), 3.92 (dd, J = 10.2, 4.4 Hz, 1H), 3.82 (d, J = 10.3 Hz, 1H), 3.75 (d, J = 4.9 Hz, 3H), 1.47 (s, 1H), 1.07 (s, 9H), 1.03 (s, 3H), 0.98 (s, 1H), 0.87 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 171.5, 168.6, 161.6, 92.4, 60.8, 59.4, 59.1, 52.3, 47.7, 47.0, 36.4, 32.7, 30.1, 27.2, 26.3, 26.1, 26.0, 25.6, 19.4, 12.3; LSMS (ESI, m/z): [M + Na]+ : 449.2.
(1R,2S,5S)-3-((S)-2-Acetamido-3,3-dimethylbutanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0] hexane-2-carboxylic acid (15a):
To a vigorously stirring solution of amide derivative 15a (40 mg, 0.12 mmol) in a 1:3 mixture of THF/H2O (0.15 M) at 0 °C was added LiOH·H2O (26 mg, 0.62 mmol). After 1 h, the solvent evaporated under reduced pressure. The reaction mixture was diluted with ethyl acetate and neutralized with 1 N HCl. The solution was extracted with EtOAc (3 × 10 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give the corresponding acid 15a (30 mg, 78%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 6.67 (d, J = 9.5 Hz, 1H), 4.60 (d, J = 9.6 Hz, 1H), 4.42 (d, J = 3.4 Hz, 1H), 3.99 – 3.87 (m, 2H), 1.99 (s, 3H), 1.61 (dd, J = 13.7, 7.5 Hz, 1H), 1.05 (s, 3H), 1.01 (s, 9H), 0.94 (s, 1H), 0.89 (s, 3H).
(1R,2S,5S)-3-((S)-2-Benzamido-3,3-dimethylbutanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0] hexane-2-carboxylic acid (15b):
Following the ester hydrolysis procedure described for the preparation of carboxylic acid 15a using ester 14b (75 mg, 0.19 mmol) afforded the acid 15b (44 mg, 61%) as a white fluffy solid. 1H NMR (400 MHz, CDCl3) δ 7.77 (d, J = 7.6 Hz, 2H), 7.47 (d, J = 7.3 Hz, 1H), 7.40 (t, J = 7.6 Hz, 2H), 7.07 (d, J = 9.7 Hz, 1H), 4.84 (d, J = 9.5 Hz, 1H), 4.44 (s, 1H), 4.05 (d, J = 10.5 Hz, 1H), 3.93 (dd, J = 10.4, 5.1 Hz, 1H), 1.50 (d, J = 7.2 Hz, 1H), 1.43 (t, J = 6.2 Hz, 1H), 1.07 (s, 9H), 1.01 (s, 3H), 0.86 (s, 3H).
(1R,2S,5S)-3-((S)-3,3-Dimethyl-2-pivalamidobutanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0] hexane-2-carboxylic acid (15c):
Following the procedure described for the preparation of carboxylic acid 15a using ester 14c (65 mg, 0.18 mmol) afforded the acid 15c (60 mg, 95%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.50 (s, 1H), 6.27 (d, J = 9.6 Hz, 1H), 4.61 (d, J = 9.7 Hz, 1H), 4.44 (s, 1H), 4.02 (d, J = 10.5 Hz, 1H), 3.86 (dd, J = 10.5, 5.4 Hz, 1H), 1.47 (dd, J = 7.5, 5.2 Hz, 1H), 1.29 (ddd, J = 13.4, 6.9, 1.6 Hz, 1H), 1.17 (d, J = 3.2 Hz, 9H), 1.04 (d, J = 2.4 Hz, 4H), 1.00 (s, 9H), 0.84 (s, 3H).
(1R,2S,5S)-3-((S)-3,3-Dimethyl-2-(2,2,2-trifluoroacetamido)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylic acid (15d):
Following the ester hydrolysis procedure described for the preparation of carboxylic acid 15a using ester 14d (600 mg, 1.59 mmol) afforded the crude compound 15d (481 mg, 83%) as a white solid. The crude residue was crystallized using a 1:4 mixture of hexanes and THF. [α]D20 −73.14 (c 0.458, MeOH). 1H NMR (400 MHz, CDCl3) δ 9.38 (bs, 1H), 7.57 (d, J = 9.5 Hz, 1H), 4.62 (d, J = 9.6 Hz, 1H), 4.45 (s, 1H), 3.93 (dd, J = 10.4, 5.1 Hz, 1H), 3.87 (d, J = 10.4 Hz, 1H), 1.58 (d, J = 7.6 Hz, 1H), 1.54–1.25 (m, 1H), 1.06 (s, 3H), 1.04 (s, 9H), 0.88 (s, 3H).
(1R,2S,5S)-3-((S)-3,3-Dimethyl-2-(2,2,2-trichloroacetamido)-butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxylic acid (15e):
Following the procedure described for the preparation of carboxylic acid 15a using ester 14e (50 mg, 0.12 mmol) afforded the acid 15e (45 mg, 94%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 10.64 (s, 1H), 7.39 (d, J = 9.2 Hz, 1H), 4.52 – 4.48 (m, 2H), 3.91 – 3.89 (m, 2H), 1.60 (d, J = 7.5 Hz, 1H), 1.54 – 1.49 (m, 1H), 1.05 (d, J = 3.6 Hz, 3H), 0.88 (s, 9H), 0.87 (s, 3H).
(1R,2S,5S)-N-((S)-1-Cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)-3-((S)-3,3-dimethyl-2-(2,2,2-trifluoroacetamido)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (1):
To a solution of tert-butyl ((S)-1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)carbamate 7 (318 mg, 1.26 mmol) in CH2Cl2 (8.3 mL) at 0 °C was added TFA (5.0 mL) and the solution was stirred for 2 h at 23 °C. After evaporating the solvent under reduced pressure, the corresponding deprotected lactam residue (192 mg, 1.26 mmol) was coupled to the acid 15d (457 mg, 1.26 mmol) using the coupling agent HATU (573 mg, 1.51 mmol) in the presence of N-methylmorphine (0.352 mL, 2.5 mmol) in DMF (6.2 mL) at 0 °C. After 10 minutes, the ice bath was removed, and the reaction mixture was stirred at 23 °C for 12 h. The solvent was then evaporated in vacuo, and the residue was dissolved in CH2Cl2. The organic layer was washed with water and brine, dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (1–2% MeOH/CH2Cl2) to give nirmatrelvir 1 (460 mg, 73%) as white fluffy solid. [α]D20 −74.36 (c 1.13, CHCl3). 1H NMR (400 MHz, DMSO-d6) δ 9.38 (d, J = 8.4 Hz, 1H), 9.00 (d, J = 8.5 Hz, 1H), 7.64 (s, 1H), 4.95 (ddd, J = 10.9, 8.5, 5.1 Hz, 1H), 4.39 (d, J = 8.6 Hz, 1H), 4.14 (s, 1H), 3.89 (dd, J = 10.4, 5.5 Hz, 1H), 3.67 (d, J = 10.4 Hz, 1H), 3.12 (t, J = 9.0 Hz, 1H), 3.02 (td, J = 9.3, 7.0 Hz, 1H), 2.41 – 2.34 (m, 1H), 2.11 (dddd, J = 20.8, 12.2, 8.7, 3.8 Hz, 2H), 1.70 (ddt, J = 17.1, 9.0, 4.3 Hz, 2H), 1.54 (dd, J = 7.6, 5.3 Hz, 1H), 1.31 (s, 1H), 1.01 (s, 3H), 0.96 (s, 9H), 0.83 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 177.8, 171.0, 167.8, 157.3 (q, J = 36.7, 36.2 Hz), 119.9, 117.7 – 114.5 (m), 60.4, 58.5, 48.0, 38.1, 37.1, 34.9, 34.5, 30.6, 27.7, 27.2, 26.1, 19.2, 12.7; 19F NMR (376 MHz, DMSO-d6) δ −74.5; HRMS (ESI/Q-TOF) m/z: [M + H]+ calcd for C23H33F3N5O4 500.2479, found: 500.2480.
(1R,2S,5S)-3-((S)-2-Acetamido-3,3-dimethylbutanoyl)-N-((S)-1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (5a):
Compound 5a was prepared in two steps in 21% yield by boc-deprotection of lactam residue 7 (25 mg, 0.097 mmol) followed by coupling to the acid 15a (30 mg, 0.097 mmol) utilizing the same procedure as described for compound 1.1H NMR (400 MHz, DMSO-d6) δ 8.95 (d, J = 8.0 Hz, 1H), 7.85 (d, J = 8.0 Hz, 1H), 7.63 (s, 1H), 4.97 – 4.91 (m, 1H), 4.28 (d, J = 8.0 Hz, 1H), 4.09 (s, 1H), 3.86 – 3.76 (m, 2H), 3.12 (t, J = 8.0 Hz, 1H), 3.05 – 2.98 (m, 1H), 2.45 – 2.36 (m, 1H), 2.17 – 2.07 (m, 1H), 1.81 (s, 3H), 1.73 – 1.64 (m, 2H), 1.52 (q, J = 4.0 Hz, 1H), 1.27 – 1.21 (m, 2H), 1.00 (s, 3H), 0.91 (s, 9H), 0.84 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 178.0, 171.4, 169.93, 169.86, 120.2, 60.3, 57.4, 47.9, 38.2, 37.2, 34.7, 34.6, 30.9, 28.9, 28.0, 27.3, 26.9, 26.3, 22.5, 19.3, 13.1; HRMS (ESI): m/z calcd for C23H35N5O4 [M+H]+ 446.2767 found 446.2763.
(1R,2S,5S)-3-((S)-2-Benzamido-3,3-dimethylbutanoyl)-N-((S)-1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (5b):
Compound 5b was prepared in two steps in 53% yield by boc-deprotection of lactam residue 7 (30 mg, 0.12 mmol) followed by coupling to the acid 15b (44 mg, 0.12 mmol) utilizing the same procedure as described for compound 1. 1H NMR (400 MHz, DMSO-d6) δ 8.97 (d, J = 8.5 Hz, 1H), 8.05 (d, J = 8.6 Hz, 1H), 7.81 – 7.76 (m, 2H), 7.64 (s, 1H), 7.49 (t, J = 7.3 Hz, 1H), 7.40 (t, J = 7.5 Hz, 2H), 5.01 – 4.93 (m, 1H), 4.55 (d, J = 8.7 Hz, 1H), 4.14 (s, 1H), 3.91 (d, J = 3.2 Hz, 2H), 3.13 (t, J = 9.1 Hz, 1H), 3.05 (dd, J = 9.4, 7.0 Hz, 1H), 2.45 – 2.35 (m, 1H), 2.12 (ddt, J = 21.5, 11.2, 5.4 Hz, 2H), 1.69 (tdd, J = 12.6, 10.5, 10.1, 4.6 Hz, 2H), 1.55 (dt, J = 6.7, 3.0 Hz, 1H), 1.28 (d, J = 7.6 Hz, 1H), 1.00 (s, 12H), 0.85 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 177.9, 171.3, 169.5, 167.5, 134.4, 131.6, 128.4, 128.1, 120.0, 60.3, 58.1, 47.9, 38.1, 37.1, 34.9, 34.5, 30.7, 27.8, 27.2, 26.9, 26.2, 19.2, 12.9; HRMS (ESI/Q-TOF) m/z: [M + Na]+ calcd for C28H37N5O4Na 530.2737, found: 530.2739.
(1R,2S,5S)-N-((S)-1-Cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)-3-((S)-3,3-dimethyl-2-pivalamidobutanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (5c):
Compound 5c was prepared in two steps in 48% yield by boc-deprotection of lactam residue 7 (32 mg, 0.13 mmol) followed by coupling to the acid 15c (45 mg, 0.13 mmol) utilizing the same procedure as described for compound 1. 1H NMR (400 MHz, DMSO-d6) δ 8.93 (d, J = 8.5 Hz, 1H), 7.65 (s, 1H), 6.85 (d, J = 9.3 Hz, 1H), 4.94 (ddd, J = 10.8, 8.4, 5.2 Hz, 1H), 4.42 (d, J = 9.3 Hz, 1H), 4.10 (s, 1H), 3.85 (dd, J = 10.3, 5.4 Hz, 1H), 3.71 (d, J = 10.4 Hz, 1H), 3.12 (t, J = 9.2 Hz, 1H), 3.03 (td, J = 9.3, 7.0 Hz, 1H), 2.39 (dd, J = 9.6, 3.9 Hz, 1H), 2.17 – 2.02 (m, 2H), 1.69 (ddt, J = 12.3, 9.8, 6.1 Hz, 2H), 1.51 (dd, J = 7.6, 5.2 Hz, 1H), 1.27 (d, J = 7.6 Hz, 1H), 1.06 (s, 9H), 0.99 (s, 3H), 0.90 (s, 9H), 0.79 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 177.9, 177.8, 171.2, 169.6, 120.0, 60.1, 56.5, 47.7, 38.5, 38.1, 37.1, 34.9, 34.4, 30.6, 27.6, 27.2, 26.7, 26.1, 19.1, 12.7; HRMS (ESI/LTQ) m/z: [M + H]+ calcd for C26H42N5O4 488.32366, found: 488.32375.
(1R,2S,5S)-N-((S)-1-Cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)-3-((S)-3,3-dimethyl-2-(2,2,2-trichloroacetamido)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (5d):
Compound 5d was prepared in two steps in 52% yield by boc-deprotection of lactam residue 7 (25 mg, 0.10 mmol) followed by coupling to the acid 15e (41 mg, 0.10 mmol) utilizing the same procedure as described for compound 1. 1H NMR (400 MHz, DMSO-d6) δ 9.00 (d, J = 8.5 Hz, 1H), 8.18 (d, J = 8.8 Hz, 1H), 7.66 (s, 1H), 4.95 (dd, J = 5.4, 2.2 Hz, 1H), 4.37 (d, J = 8.8 Hz, 1H), 4.16 (s, 1H), 3.91 (dd, J = 10.4, 5.5 Hz, 1H), 3.66 (d, J = 10.5 Hz, 1H), 3.16 – 3.08 (m, 1H), 3.03 (td, J = 9.3, 6.9 Hz, 1H), 2.37 (dd, J = 9.3, 4.6 Hz, 1H), 2.16 – 2.02 (m, 2H), 1.70 (ddt, J = 12.9, 10.0, 6.8 Hz, 2H), 1.56 (dd, J = 7.6, 5.3 Hz, 1H), 1.32 (s, 1H), 1.22 (d, J = 6.5 Hz, 1H), 1.01 (s, 3H), 0.97 (s, 9H), 0.83 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 177.8, 171.0, 167.8, 161.9, 119.9, 92.8, 60.5, 59.3, 48.0, 38.2, 37.1, 35.7, 34.4, 30.6, 27.6, 27.2, 26.4, 26.1, 19.2, 12.8; HRMS (ESI/LTQ) m/z: [M + H]+ calcd for C23H33Cl3N5O4 548.1598, found: 548.1604.
(1R,2S,5S)-N-((S)-1-Cyano-2-((S)-2-oxopiperidin-3-yl)ethyl)-3-((S)-3,3-dimethyl-2-(2,2,2-trifluoroacetamido)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (5e):
Compound 5e was prepared in two steps in 64% yield by boc-deprotection of lactam residue 7 (94 mg, 0.35 mmol) followed by coupling to the acid 15d (127 mg, 0.35 mmol) utilizing the same procedure as described for compound 1. 1H NMR (400 MHz, DMSO-d6) δ 9.38 (d, J = 8.4 Hz, 1H), 8.97 (d, J = 8.2 Hz, 1H), 4.99 (ddd, J = 10.4, 8.2, 5.7 Hz, 1H), 4.39 (d, J = 8.5 Hz, 1H), 4.15 (s, 1H), 3.88 (dd, J = 10.4, 5.5 Hz, 1H), 3.66 (d, J = 10.5 Hz, 1H), 3.06 (dt, J = 8.2, 3.6 Hz, 2H), 2.35 – 2.17 (m, 2H), 1.88 – 1.78 (m, 1H), 1.71 (tdd, J = 18.2, 8.7, 5.2 Hz, 2H), 1.54 (dd, J = 7.7, 5.3 Hz, 2H), 1.40 – 1.31 (m, 1H), 1.27 (d, J = 7.6 Hz, 1H), 1.00 (s, 3H), 0.97 (s, 9H), 0.82 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 172.2, 171.0, 167.7, 157.4, 120.1, 117.6, 60.3, 58.5, 47.9, 41.5, 38.2, 37.0, 34.9, 34.7, 30.7, 27.6, 26.6, 26.1, 21.6, 19.2, 12.7; HRMS (ESI/LTQ) m/z: [M + H]+ calcd for C24H35F3N5O4 514.26409, found: 514.26396.
(1R,2S,5S)-N-((S)-1-Cyano-2-((S)-2-oxopiperidin-3-yl)ethyl)-3-((S)-3,3-dimethyl-2-(2,2,2-trichloroacetamido)butanoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (5f):
Compound 5f was prepared in two steps in 37% yield by boc-deprotection of lactam residue 8 (18 mg, 0.07 mmol) followed by coupling to the acid 15e (28 mg, 0.07 mmol) utilizing the same procedure as described for compound 1. 1H NMR (400 MHz, DMSO-d6) δ 8.97 (d, J = 8.2 Hz, 1H), 8.16 (d, J = 8.8 Hz, 1H), 7.49 (d, J = 2.8 Hz, 1H), 4.99 (ddd, J = 10.4, 8.2, 6.0 Hz, 1H), 4.38 (d, J = 8.8 Hz, 1H), 4.17 (s, 1H), 3.91 (dd, J = 10.5, 5.5 Hz, 1H), 3.65 (d, J = 10.4 Hz, 1H), 3.11 – 3.02 (m, 2H), 2.25 (dddd, J = 33.2, 14.5, 10.7, 5.7 Hz, 2H), 1.88 – 1.79 (m, 1H), 1.71 (dddd, J = 17.8, 13.3, 8.7, 5.3 Hz, 2H), 1.55 (dd, J = 7.6, 5.2 Hz, 2H), 1.37 (dd, J = 13.9, 11.1 Hz, 2H), 1.29 (d, J = 7.6 Hz, 1H), 1.01 (s, 3H), 0.98 (s, 9H), 0.83 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 172.3, 170.9, 167.8, 161.9, 120.1, 92.8, 60.4, 59.3, 47.9, 41.5, 38.3, 37.0, 35.8, 34.7, 30.7, 27.5, 26.5, 26.1, 21.6, 19.2, 12.8; HRMS (ESI/Q-TOF) m/z: [M + H]+ calcd for C24H35Cl3N5O4 562.1749, found: 562.1764.
Tert-butyl ((S)-1-((1R,2S,5S)-2-(((S)-1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)carbamoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexan-3-yl)-3,3-dimethyl-1-oxobutan-2-yl)carbamate (5g):
To a solution of lactam 7 (37 mg, 0.15 mmol) in CH2Cl2 (1.0 mL) at 0 °C was added TFA (0.6 mL) and the solution was stirred for 1 h at 23 °C. After evaporating the solvent under reduced pressure, the corresponding deprotected lactam residue (22 mg, 0.15 mmol) was coupled to the acid 6a (54 mg, 0.15 mmol) using EDC·HCl (34 mg, 0.18 mmol), HOBt (27 mg, 0.18 mmol) and N-methylmorphine (24 μL, 0.22 mmol) in DMF (1.8 mL) at 0 °C. After 10 minutes, the ice bath was removed, and the reaction mixture was stirred at 23 °C for 12 h. The solvent was then evaporated in vacuo, and the residue was dissolved in CH2Cl2. The organic layer was washed with water and brine, dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (1–2% MeOH/ CH2Cl2) to give the amide 5g (46 mg, 62% over 2-steps) as white fluffy solid. 1H NMR (400 MHz, DMSO-d6) δ 8.96 (d, J = 8.6 Hz, 1H), 7.63 (s, 1H), 6.64 (d, J = 9.1 Hz, 1H), 4.93 (ddd, J = 10.9, 8.5, 5.1 Hz, 1H), 4.13 (s, 1H), 3.98 (d, J = 9.2 Hz, 1H), 3.89 – 3.77 (m, 2H), 3.14 – 2.96 (m, 2H), 2.38 (dt, J = 10.2, 4.9 Hz, 1H), 2.18 – 2.01 (m, 2H), 1.76 – 1.60 (m, 2H), 1.51 (dd, J = 7.7, 5.1 Hz, 1H), 1.32 (s, 9H), 1.25 (d, J = 7.6 Hz, 1H), 1.00 (s, 3H), 0.89 (s, 9H), 0.85 (s, 3H); 13C NMR (100 MHz, DMSO-d6) δ 177.8, 171.3, 170.3, 156.3, 120.0, 78.5, 60.1, 59.1, 47.8, 38.1, 37.0, 34.4, 34.3, 30.6, 28.4, 27.8, 27.2, 26.7, 26.2, 19.2, 12.8; HRMS (ESI/LTQ) m/z: [M + H]+ calcd for C26H42N5O5 504.31857, found: 504.31869.
(1R,2S,5S)-N-((S)-1-Cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)-3-((S)-3,3-dimethyl-2-(2,2,2-trifluoroacetamido)butanoyl)-6,6-dimethyl-N-(2,2,2-trifluoroacetyl)-3-azabicyclo[3.1.0] hexane-2-carboxamide (5h):
Following the procedure described for the preparation of compound 14d with trifluoloroacetic anhydride (14 mg, 0.1 mmol) and ZnCl2 (5 mol%) afforded the compound 5h (10 mg, 85%) as a white sticky solid. 1H NMR (400 MHz, DMSO-d6) δ 11.69 (s, 1H), 9.41 (d, J = 8.3 Hz, 1H), 7.64 (s, 1H), 4.87 (s, 1H), 4.43 (d, J = 8.4 Hz, 1H), 3.92 (dd, J = 10.6, 5.2 Hz, 1H), 3.82 (d, J = 10.6 Hz, 1H), 3.13 – 3.05 (m, 2H), 2.74 – 2.68 (m, 1H), 2.42 (dt, J = 9.3, 4.6 Hz, 1H), 2.25 (dd, J = 14.6, 10.3 Hz, 1H), 2.11 – 1.99 (m, 1H), 1.75 – 1.58 (m, 2H), 1.45 (d, J = 7.5 Hz, 1H), 1.00 (s, 4H), 0.94 (s, 9H), 0.86 (s, 3H); 13C NMR (200 MHz, DMSO-d6) δ 178.0, 168.2, 160.3, 157.4, 157.4, 156.5, 156.3, 136.0, 131.0, 127.8, 116.9, 116.5, 115.1, 114.2, 79.5, 79.3, 79.1, 59.0, 54.7, 47.8, 35.0, 31.2, 29.4, 27.5, 26.6, 26.5, 26.4, 26.1, 22.5, 19.4, 12.4; HRMS (ESI/LTQ) m/z: [M + H]+ calcd for C25H32F6N5O5 596.2307, found: 596.2321.
Benzyl ((S)-1-((1R,2S,5S)-2-(((S)-1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)carbamoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexan-3-yl)-3,3-dimethyl-1-oxobutan-2-yl)carbamate (5i):
To a vigorously stirring solution of amide derivative 6b (65 mg, 0.16 mmol) in a 1:3 mixture of THF/H2O (3.0 mL) at 0 °C was added LiOH·H2O (20 mg, 0.5 mmol). After 1 h, the solvent evaporated under reduced pressure. The reaction mixture was diluted with ethyl acetate and neutralized with 1 N HCl. The solution was extracted with EtOAc (3 × 5 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give the crude acid (58 mg, 92%) as a white solid.
To a solution of tert-butyl((S)-1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)carbamate 7 (20 mg, 0.08 mmol) in CH2Cl2 (1.0 mL) at 0 °C was added TFA (0.2 mL) and the solution was stirred for 2 h at 23 °C. After evaporating the solvent under reduced pressure, the corresponding deprotected lactam residue (10 mg, 0.064 mmol) was coupled to the above crude acid (26 mg, 0.064 mmol) using the coupling agent HATU (38 mg, 0.1 mmol) and DIPEA (0.13 ml, 0.65 mmol) in DMF (2.0 mL) at 0 °C. After 10 minutes, the ice bath was removed, and the reaction mixture was stirred at 23 °C for 12 h. The solvent was then evaporated in vacuo, and the residue was dissolved in CH2Cl2. The organic layer was washed with water and brine, dried over Na2SO4, concentrated under reduced pressure and the residue was purified by column chromatography (1–2% MeOH/ CH2Cl2) to give compound 5i (14 mg, 40%) as white fluffy solid. 1H NMR (400 MHz, CDCl3) δ 8.08 (d, J = 8.0 Hz, 1H), 7.28 – 7.38 (m, 6H), 5.78 (s, 1H), 5.43 (d, J = 8.5 Hz, 2H), 5.05 (d, J = 3.22 Hz, 2H), 4.88–4.92 (m, 1H), 4.26 (s, 2H), 3.90 (s, 1H), 3.29–3.36 (m, 2H), 2.51–2.57 (m, 1H), 2.30–2.37 (m, 2H), 1.86–2.01 (m, 1H), 1.54–1.83 (m, 1H), 1.35–1.37 (t, J = 2.5 Hz, 1H), 1.05 (s, 3H), 0.96 (s, 9H), 0.85 (s, 3H); 13C NMR (200 MHz, CDCl3) δ 171.5, 171.3, 170.2, 167.6, 156.3, 136.4, 128.52, 128.49, 128.0, 118.3, 67.0, 60.6, 59.3, 48.3, 40.4, 37.6, 35.3, 29.7, 27.9, 26.4, 26.2, 19.3, 14.1, 13.4, 12.7; HRMS (ESI/LTQ) m/z. [M+H]+ calcd for C29H39N5O5: 538.3029; found 538.3020.
Pyridin-2-ylmethyl((2S)-1-((1R,2S,5S)-2-((1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl) carbamoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexan-3-yl)-3,3-dimethyl-1-oxobutan-2-yl)carbamate (5j):
(S)-2-Amino-3-((S)-2-oxopyrrolidin-3-yl)propanenitrile (12 mg, 0.079 mmol) was coupled with the corresponding acid (32 mg, 0.079 mmol) utilizing the same procedure as described for compound 5i to afford the compound 5j (15 mg, 35%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.97 (d, J = 8.5 Hz, 1H), 8.50 – 8.49 (m, 1H), 7.77 (td, J = 7.7, 1.8 Hz, 1H), 7.64 (s, 1H), 7.47 (d, J = 8.9 Hz, 1H), 7.32 – 7.27 (m, 2H), 5.03 (q, J = 13.8 Hz, 2H), 4.94 (ddd, J = 10.9, 8.5, 5.2 Hz, 1H), 4.13 (s, 1H), 4.06 (d, J = 8.9 Hz, 1H), 3.84 (dd, J = 10.2, 5.4 Hz, 1H), 3.76 (d, J = 10.3 Hz, 1H), 3.11 (t, J = 9.1 Hz, 1H), 3.02 (td, J = 9.3, 7.0 Hz, 1H), 2.39 (dt, J = 10.1, 4.9 Hz, 1H), 2.17 – 2.03 (m, 2H), 1.73 – 1.64 (m, 1H), 1.51 (dd, J = 7.6, 5.2 Hz, 1H), 1.26 (d, J = 7.5 Hz, 2H), 0.99 (s, 3H), 0.93 (s, 9H), 0.79 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 178.0, 171.4, 169.9, 157.2, 156.8, 149.5, 137.3, 123.2, 121.4, 120.1, 66.7, 60.4, 59.8, 48.0, 38.3, 37.2, 34.8, 34.6, 30.8, 27.9, 27.4, 26.8, 26.3, 19.3, 13.0; HRMS (ESI): m/z calcd for C28H38N6O5 [M+H]+ 539.2981 found 539.2975.
Pyridin-3-ylmethyl((2S)-1-((1R,2S,5S)-2-((1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl) carbamoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexan-3-yl)-3,3-dimethyl-1-oxobutan-2-yl)carbamate (5k):
(S)-2-Amino-3-((S)-2-oxopyrrolidin-3-yl)propanenitrile (13 mg, 0.081 mmol) was coupled with the corresponding acid (33 mg, 0.081 mmol) utilizing the same procedure as described for compound 5i to afford the compound 5k (14 mg, 32%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.96 (d, J = 8.5 Hz, 1H), 8.51 (d, J = 15.1 Hz, 2H), 7.71 (d, J = 8.1 Hz, 1H), 7.63 (s, 1H), 7.41 – 7.34 (m, 2H), 5.09 – 5.00 (m, 2H), 4.95 (d, J = 13.5 Hz, 1H), 4.10 (s, 1H), 4.04 (d, J = 8.7 Hz, 1H), 3.82 (d, J = 6.0 Hz, 1H), 3.76 (d, J = 10.4 Hz, 1H), 3.11 (t, J = 9.1 Hz, 1H), 3.02 (t, J = 8.4 Hz, 1H), 2.38 (d, J = 9.9 Hz, 1H), 2.09 (ddd, J = 24.7, 13.1, 5.4 Hz, 2H), 1.67 (d, J = 10.5 Hz, 1H), 1.51 (t, J = 6.5 Hz, 1H), 1.24 (dd, J = 11.0, 7.5 Hz, 2H), 1.00 (s, 3H), 0.91 (s, 9H), 0.80 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 178.0, 171.4, 169.9, 156.8, 149.5, 136.0, 131.1, 123.9, 120.1, 63.7, 60.3, 59.8, 48.0, 38.2, 37.2, 34.7, 34.6, 30.8, 28.0, 27.4, 26.8, 26.3, 19.3, 13.0; HRMS (ESI): m/z calcd for C28H38N6O5 [M+H]+ 539.2981 found 539.2976.
Pyridin-4-ylmethyl((2S)-1-((1R,2S,5S)-2-((1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl) carbamoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexan-3-yl)-3,3-dimethyl-1-oxobutan-2-yl)carbamate (5l):
(S)-2-Amino-3-((S)-2-oxopyrrolidin-3-yl)propanenitrile (12 mg, 0.074 mmol) was coupled with the corresponding acid (30 mg, 0.074 mmol) utilizing the same procedure as described for compound 5i to afford the compound 5l (14 mg, 35%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.97 (d, J = 8.5 Hz, 1H), 8.52 (s, 2H), 7.64 (s, 1H), 7.52 (d, J = 8.9 Hz, 1H), 7.33 – 7.22 (m, 2H), 5.12 – 5.00 (m, 2H), 4.94 (ddd, J = 10.9, 8.5, 5.2 Hz, 1H), 4.12 (s, 1H), 4.03 (dd, J = 18.6, 8.0 Hz, 1H), 3.88 – 3.69 (m, 2H), 3.14 – 3.01 (m, 2H), 2.14 – 2.04 (m, 2H), 1.70 – 1.66 (m, 1H), 1.51 (dd, J = 7.7, 5.3 Hz, 1H), 1.27 – 1.23 (m, 2H), 0.98 (s, 3H), 0.93 (s, 9H), 0.77 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 178.0, 171.4, 169.9, 156.7, 150.0, 146.9, 121.9, 120.1, 64.2, 60.4, 59.8, 48.0, 38.3, 37.2, 34.8, 34.6, 30.8, 27.9, 27.4, 26.8, 26.3, 19.3, 12.9; LRMS-ESI (m/z) [M+H]+ 539.4.
(1R,2S,5S)-3-((S)-2-(3-(Tert-butyl)ureido)-3,3-dimethylbutanoyl)-N-((S)-1-cyano-2-((S)-2-oxopyrrolidin-3-yl)ethyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexane-2-carboxamide (5m):
(S)-2-Amino-3-((S)-2-oxopyrrolidin-3-yl)propanenitrile (13 mg, 0.087 mmol) was coupled with the corresponding acid (32 mg, 0.087 mmol) utilizing the same procedure as described for compound 5i to afford the compound 5m (14 mg, 32%) as a white solid. 1H NMR (400 MHz, CDCl3) δ 8.13 (d, J = 8.0 Hz, 1H), 6.35 (s, 1H), 5.17 (s, 1H), 5.06 (q, J = 5.96 Hz, 1H), 4.97 (s, 1H), 4.33 (d, J = 9.54 Hz, 1H), 4.28 (s, 1H), 4.12 (d, J = 10.14 Hz, 1H), 3.89–3.92 (m, 1H), 3.27–3.33 (m, 1H), 2.50–2.54 (m, 1H), 2.30–2.40 (m, 2H), 1.93–1.97 (m, 1H), 1.80–1.84 (m, 1H), 1.55 (t, J = 2.54 Hz, 1H), 1.43–1.45 (m, 1H), 1.32–1.34 (t, J = 2.12 Hz, 1H), 1.24 (s, 9H), 1.03 (s, 3H), 0.96 (s, 9H), 0.92 (s, 3H); 13C NMR (100 MHz, CDCl3) δ 179.1, 173.1, 171.2, 157.5, 119.4, 60.5, 58.0, 50.2, 48.4, 40.4, 38.9, 37.6, 34.7, 34.5, 30.2, 29.4, 28.0, 26.7, 26.3, 19.3, 12.6; HRMS (ESI): m/z calcd for C26H42N6O4 [M+H]+ 503.3345; found 503.3337.
Pyridin-3-ylmethyl((S)-1-((1R,2S,5S)-2-(((S)-1-cyano-2-((S)-2-oxopiperidin-3-yl)ethyl) carbamoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexan-3-yl)-3,3-dimethyl-1-oxobutan-2-yl)carbamate (5n):
(S)-2-Amino-3-((S)-2-oxopiperidin-3-yl)propanenitrile (12 mg, 0.073 mmol) was coupled with the corresponding acid (27 mg, 0.073 mmol) utilizing the same procedure as described for compound 5i to afford the compound 5n (15 mg, 34%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.93 (d, J = 8.0 Hz, 1H), 8.54 (s, 1H), 8.50 (d, J = 4.0 Hz, 1H), 7.74 – 7.71 (m, 1H), 7.47 (s, 1H), 7.39 – 7.35 (m, 2H), 5.09 (d, J = 12.0 Hz, 1H), 5.00 – 4.94 (m, 2H), 4.12 (s, 1H), 4.04 (d, J = 8.0 Hz, 1H), 3.85 – 3.75 (m, 2H), 3.05 (t, J = 4.0 Hz, 2H), 2.34 – 2.21 (m, 2H), 1.84 – 80 (m, 1H), 1.75 – 1.64 (m, 1H), 1.51 (t, J = 4.0 Hz, 2H), 1.38 – 1.34 (m, 1H), 1.23 (d, J = 8.0 Hz, 2H), 0.99 (s, 3H), 0.92 (s, 9H), 0.79 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 172.4, 171.4, 169.8, 156.8, 149.4, 136.1, 133.2, 123.9, 120.3, 63.7, 60.2, 59.8, 47.9, 41.6, 38.3, 37.1, 34.8, 34.7, 30.9, 27.8, 26.8, 26.3, 26.2, 21.8, 19.3, 12.9; HRMS (ESI): m/z calcd for C29H40N6O5 [M+H]+ 553.3138 found 553.3132.
Pyridin-4-ylmethyl((S)-1-((1R,2S,5S)-2-(((S)-1-cyano-2-((S)-2-oxopiperidin-3-yl)ethyl) carbamoyl)-6,6-dimethyl-3-azabicyclo[3.1.0]hexan-3-yl)-3,3-dimethyl-1-oxobutan-2-yl)carbamate (5o):
(S)-2-Amino-3-((S)-2-oxopiperidin-3-yl)propanenitrile (13 mg, 0.074 mmol) was coupled with the corresponding acid (30 mg, 0.074 mmol) utilizing the same procedure as described for compound 5i to afford the compound 5o (14 mg, 34%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ 8.94 (d, J = 8.2 Hz, 1H), 8.51 (s, 2H), 7.60 – 7.40 (m, 2H), 7.27 (d, J = 5.4 Hz, 2H), 5.14 – 4.91 (m, 3H), 4.13 (s, 1H), 4.06 (d, J = 8.9 Hz, 1H), 3.88 – 3.72 (m, 2H), 3.12 – 3.01 (m, 2H), 2.35 – 2.18 (m, 2H), 1.83 (dd, J = 12.8, 4.0 Hz, 1H), 1.78 – 1.63 (m, 2H), 1.55 – 1.44 (m, 2H), 1.23 (d, J = 7.1 Hz, 2H), 0.98 (s, 3H), 0.94 (s, 9H), 0.76 (s, 3H); 13C NMR (125 MHz, DMSO-d6) δ 172.4, 171.3, 169.8, 156.7, 150.0, 146.8, 121.9, 120.3, 64.2, 60.2, 59.8, 47.9, 41.6, 38.3, 37.1, 34.84, 34.79, 30.9, 27.8, 26.9, 26.3, 26.2, 21.8, 19.3, 12.9; HRMS (ESI): m/z calcd for C29H40N6O5 [M+H]+ 553.3138 found 553.3131.
Expression and Purification of SARS-CoV-2 main protease
The gene encoding the SARS-CoV-2 main protease (Mpro), also known as 3C like protease (3CLpro) was obtained from the polyprotein sequence (Accession #: MN908947). The expression construct was designed to contain an N-terminal hexa-histidine tag. This was followed by polyprotein residues 3259–3569 to reflect the sequence of the 3CLpro enzyme in addition to the nsp4/5 autocleavage site. The 3CLpro nucleotide sequence was codon optimized for expression in E. Coli, synthesized and subcloned into a pET11a vector by BioBasic. This expression construct produced a protein with its authentic N- and C-termini.
This plasmid containing the SARS-CoV-2 3CLpro construct was transformed into E. coli BL21 (DE3) cells by electroporation. A single colony of the transformed cells was used to inoculate 150 mL of Super LB media (3 g monobasic potassium phosphate, 6 g dibasic sodium phosphate, 20 g tryptone, 5 g yeast extract, 5 g NaCl per 1 L of water, 100 μg/mL Carbenicillin, pH adjusted to 7.20 using 10 M NaOH). This preculture was incubated overnight for 12 hours at 37 °C with shaking. The expression culture consisted of 500 mL Super LB media supplemented with 100 μg/mL Carbenicillin, 12.5 mL 8% lactose, 5 mL 60% glycerol, 2.5 mL 10% glucose, and 12.5 mL of preculture that was incubated at 25 °C for 24 hours. The cells were harvested via centrifugation at 16,800 x g for 40 minutes to yield 7.2 g L−1 of cells. The cell pellet was resuspended in 5 mL Lysis Buffer (25 mM HEPES, 0.05 mM EDTA, 5 mM β-ME, 1 mg/mL lysozyme) per 1 g of pelleted cells using a manual homogenizer. Homogenized cell suspension was sonicated for a total of 10 minutes at an amplitude of 60% for periods of 10 s punctuated by 20 s delays using a Branson digital sonifier. The lysate produced was clarified by centrifugation for 16,080 x g for 45 minutes at 4 °C.
Clarified lysate was injected onto a 60 mL Q-Sepharose Fast Flow (GE Healthcare) strong anion-exchange column equilibrated in Buffer A (25 mM HEPES pH 7.50, 0.05 mM EDTA, 5 mM β-ME). Protein was recovered from the flow-through and an elution phase to ~45 % Buffer B (50 mM HEPES pH 7.5, 1 M Ammonium sulfate, 0.05 mM EDTA, 5 mM β-ME) over ~5 column volumes. Protein fractions were pooled based upon those with the highest specific activity as well as by visual inspection of purity using SDS-PAGE gels. Solid Ammonium sulfate was gradually added to the protein pool to a final concentration of 1 M while stirring on ice. Precipitation was removed via centrifugation at 16,000 x g for 45 minutes at 4 °C. Supernatant was checked for activity prior to injection onto a 60 mL Phenyl Sepharose 6 Fast Flow Hi Sub (GE Healthcare) hydrophobic interaction column that had been equilibrated in Buffer B. Protein was eluted using a linear gradient to 100% Buffer A over ~12 column volumes. Protein eluted at ~0.6 column volumes after reaching 100% Buffer B. Fractions containing CoVID-19 3CLpro were assessed for purity and pooled based on the criteria above. The pH of the pool was adjusted to pH 5.5 using a 1 M MES solution while stirring on ice. This protein was syringe-filtered with a 0.45 μm SFCA membrane prior to injection onto an 8 mL Mono S (GE Healthcare) equilibrated in Buffer C (20 mM MES monohydrate pH 5.50, 0.05 mM EDTA, 5 mM β-ME) for cation-exchange chromatography. Protein was eluted using a gradient to ~60% Buffer D (50 mM MES monohydrate pH 5.5, 1 M NaCl, 0.05 mM EDTA, 5 mM β-ME) over ~46 column volumes. Fractions were pooled using the same criteria. This pool was concentrated using a 10,000 MWCO spin concentrator (Millipore) before being injected onto a 300 mL HiLoad 26/60 Superdex 200 prep grade column (GE Healthcare) for size-exclusion and desalting into Buffer E (25 mM HEPES pH 7.5, 10% Glycerol, 2.5 mM DTT). Protein was pooled based on the above criteria, aliquoted, flash-frozen in liquid nitrogen, and stored at −80 °C. The protein concentration was determined by measuring absorbance at 280 nm and calculating the concentration using a molar extinction coefficient of 32,890 M−1 cm−1 determined from ProtParam (Expasy).
SARS-CoV-2 3CLpro inhibition assays
Inhibition of SARS-CoV-2 3CLpro by designed compounds was assessed similar to our previously published methods using a continuous fluorescence assay and the FRET-based substrate UIVT3 (HiLyte Fluor488™–ESATLQSGLRKAK-QXL520™-NH2) which was synthesized by Anaspec, Fremont, CA.40,44 Briefly, the assay buffer consisted of 50 mM HEPES pH 7.5, 0.1 mg/mL BSA, 0.01% Triton X-100, 2 mM DTT, 1% DMSO and a final enzyme concentration of 50 nM to 200 nM depending on inhibitory potency. Kinetic assays were performed in Costar 3694 EIA/RIA 96-well half-area, flat bottom, black polystyrene plates (Corning, Corning, NY) at 25 °C. The increase in fluorescence intensity was measured at an emission wavelength of 528 nm (20 nm bandwidth) using an excitation wavelength of 485 (bandwidth 20 nm) using a CLARIOstar Plate Reader (BMG Labtech, Cary, NC). The initial rates of the reactions were determined from the slopes of the Relative Fluorescence Units (RFU) versus time (RFU min−1). Inhibition data were collected and analyzed as described previously.40
X-ray Structure determination of inhibitor 5e (GRL-190–21) bound to SARS-CoV-2 3CLpro.
Co-crystals of SARS-CoV-2 3CLpro with 5e (GRL-190–21) were grown at 4 °C using the sitting-drop or hanging-drop vapor diffusion method as described previously.40,44 Briefly, 1 μL of ~110 μM SARS-CoV-2 3CLpro in 25 mM HEPES pH 7.5, 2.5 mM DTT containing ~150 μM 5e (GRL-190–21) was mixed with 2 μL of reservoir solution which contained a constant concentration of 3 mM DTT, 50 mM MES pH 6.0, 1% MPD, and varying concentrations of PEG-10,000 and KCl. Crystals that formed were removed using nylon loops and transferred into 3 μL of a cryo-solution containing the crystallization solution and inhibitor that was supplemented with 30% MPD. Crystals were soaked in the resulting solution for about 30 minutes and then flash-frozen in liquid nitrogen for X-ray data collection.
X-ray diffraction data were collected on crystals using Life Sciences Collaborative Access Team (LS-CAT) beamline 21-ID-F at the Advanced Photon Source, Argonne National Laboratory. X-ray data were indexed, integrated and scaled using the HKL2000 software package.45 The software package PHENIX was used for structure determination via molecular replacement and iterative rounds of structural refinement.46 Inhibitor coordinates and restraints were generated using eLBOW (PHENIX).47 Manual modeling building was performed using Coot.48 Automated structural refinement was performed using the Refine module avilalbe in PHENIX. The X-ray data collection and refinement statistics for the SARS-CoV-2:3Clpro GRL-190–21 inhibitor complex are summarized in Table 4.43 The resulting coordinates and associated reflection files are deposited in the PDB under accession number 8UND.
Supplementary Material
Highlights.
Design and synthesis of potent SARS-CoV-2 Mpro inhibitors are reported.
Inhibitors with a six-membered P1 lactam exhibited improved antiviral activity.
Inhibitors maintained potent antiviral activity against variants of concern.
An inhibitor exhibited improved antiviral profile in immunocytochemistry assays.
The X-ray crystal structure of SARS-CoV-2 Mpro inhibitor complex was determined.
Acknowledgements
The research was supported in part by grants from the National Institute of Allergy and Infectious Diseases, National Institutes of Health (A.K.G. AI150466, and A.K.G. & A.D.M., AI158649 and contract No. HHSN272201700060C). The present work was also supported by a grant for Development of Novel Drugs for Treating COVID-19 from the Intramural Research Program of National Center for Global Health and Medicine (H.M., 19A3001 and Y.T., 22A2008D), in part by Japan Agency for Medical Research and Development (AMED) (H.M., 21fk0108480). UJ was supported by NIH NIAID T32 training grant AI 148103 (Drug Discovery in Infectious Disease Training) and SB was supported by NIH NIGMS T32 training grant GM132024 (Purdue University Molecular Biophysics Training Program). The authors also wish to acknowledge support from the Purdue Institute for Cancer Research, NIH grant P30 CA023168, for use of the shared NMR and Macromolecular Crystallization and X-ray diffraction facilities available in the Biomolecular Structure Shared Resource. X-ray data were collected at the Life Sciences Collaborative Access Team (LS-CAT) beamline 21-ID-F at the Advanced Photon Source (APS), Argonne National Laboratory (ANL). The APS is a U.S. Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under Contract No. DE-AC02-06CH11357. Use of the LS-CAT Sector 21 was supported by the Michigan Economic Development Corporation and the Michigan Technology Tri-Corridor (Grant 085P1000817).
Abreviations used
- CYP
Cytochrome p450
- MPro
Main Protease
- PDB
Protein data base
- SAR
Structure-Activity Relationship
- SARS
CoV-2 Severe Acute Respiratory Syndrome Coronavirus-2
- RdRp
RNA-dependent RNA polymerase
- VOCs
Variants of concern
Footnotes
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Appendix A. Supplementary Material
Supplementary data to this article can be found online.
Full NMR spectroscopic data for all final compounds.
Methods for expression and purification of SARS-CoV-2 Mpro.
Methods for SARS-CoV-2 Mpro inhibition assay.
Methods for crystallization of SARS-CoV Mpro with inhibitors.
X-ray structural data for inhibitor 5f-bound SARS-CoV-2 MPro.
The PDB accession code for inhibitor 5f-bound SARS-CoV-2 Mpro X-ray structure has been deposited (8UND).
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.
Data Availability
X-ray crystallographic data for compound 5e has been deposited at the PDB (8UND). Spectral data, assay, and X-ray data supporting this article are available for public use.
References
- 1.Rutter H, Wolpert M, Greenhalgh T. Managing uncertainty in the covid-19 era. British. Med. Journal, 2020, 370, m3349. [DOI] [PubMed] [Google Scholar]
- 2.Wang C, Horby PW, Hayden FG, Gao GF. A novel coronavirus outbreak of global health concern. Lancet, 2020, 395, 470–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.WHO Coronavirus (COVID-19) Dashboard. https://covid19.who.int (accessed 2023–10-08).
- 4.Murray CJL. COVID-19 will continue but the end of the pandemic is near. Lancet, 2022, 399, 417–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Del Rio C, Malani PN. COVID-19 in 2022-The Beginning of the End or the End of the Beginning? JAMA, 2022, 327, 2389–2390. [DOI] [PubMed] [Google Scholar]
- 6.Pelly S, Liotta D. Potent SARS-CoV-2 Direct-Acting Antivirals Provide an Important Complement to COVID-19 Vaccines. ACS Cent. Sci, 2021, 7 (3), 396–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chen L-L, Abdullah SMU, Chan W-M, Chan BP-C, Ip JD, Chu AW-H, Lu L, Zhang X, Zhao Y, Chuang VW-M, Au AK-W, Cheng VC-C, Sridhar S, Yuen K-Y, Hung IF-N, Chan K-H, To KK-W. Contribution of Low Population Immunity to the Severe Omicron BA.2 Outbreak in Hong Kong. Nat Commun, 2022, 13 (1), 3618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lu L, Mok BWY, Chen LL, Chan JMC, Tsang OTY, Lam BHS, Chuang VWM, Chu AWH, Chan WM, Ip JD, Chan BPC, Zhang R, Yip CCY, Cheng VCC, Chan KH, Jin DY, Hung IFN, Yuen KY, Chen H, To KKW. Neutralization of Severe Acute Respiratory Syndrome Coronavirus 2 Omicron Variant by Sera From BNT162b2 or CoronaVac Vaccine Recipients. Clinical Infectious Diseases, 2022, 75 (1), e822–e826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Søgaard OS, Reekie J, Johansen IS, Nielsen H, Benfield T, Wiese L, N. B. Stærke K.; Iversen K. Fogh, Bodilsen J, Iversen M, Knudsen LS, Klastrup V, Larsen FD, Andersen SD, Hvidt AK, Andreasen SR, Madsen LW, Lindvig SO, Øvrehus A, Ostrowski SR, Abildgaard C, Matthews C, Jensen TO, Raben D, Erikstrup C, Fischer TK, Tolstrup M, Østergaard L, Lundgren J. Characteristics Associated with Serological COVID-19 Vaccine Response and Durability in an Older Population with Significant Comorbidity: The Danish Nationwide ENFORCE Study. Clinical Microbiology and Infection, 2022, 28 (8), 1126–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kannan SR, Spratt AN, Sharma K, Chand HS, Byrareddy SN, Singh K. Omicron SARS-CoV-2 variant: Unique features and their impact on pre-existing antibodies. J. Autoimmun, 2022, 126, 102779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Karim SSA, Karim QA. Omicron SARS-CoV-2 variant: A new chapter in the COVID-19 pandemic. Lancet. 2021, 398, 2126–2128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Nagu P, Parashar A, Behl T, Mehta V. CNS Implications of COVID-19: A Comprehensive Review. Reviews in the Neurosciences, 2021, 32 (2), 219–234. [DOI] [PubMed] [Google Scholar]
- 13.Sato K, Sinclair JE, Sadeghirad H, Fraser JF, Short KR, Kulasinghe A. Cardiovascular Disease in SARS-CoV-2 Infection. Clinical & Translational Immunology, 2021, 10 (9), e1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Owen DR, Allerton CMN, Anderson AS, Aschenbrenner L, Avery M, Berritt S, Boras B, Cardin RD, Carlo A, Coffman KJ, Dantonio A, Di L, Eng H, Ferre R, Gajiwala KS, Gibson SA, Greasley SE, Hurst BL, Kadar EP, Kalgutkar AS, Lee JC, Lee J, Liu W, Mason SW, Noell S, Novak JJ, Obach RS, Ogilvie K, Patel NC, Pettersson M, Rai DK, Reese MR, Sammons MF, Sathish JG, Singh RSP, Steppan CM, Stewart AE, Tuttle JB, Updyke L, Verhoest PR, Wei L, Yang Q, Zhu Y. An oral SARS-CoV-2 Mpro inhibitor clinical candidate for the treatment of COVID-19. Science, 2021, 374, 1586–1593. [DOI] [PubMed] [Google Scholar]
- 15.Lamb YN; Nirmatrelvir Plus Ritonavir: First Approval. Drugs. 2022, 82, 585–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bernal AJ., Gomes da Silva MM, Musungaie DB, Kovalchuk E, Gonzalez A, Delos Reyes V, Martín-Quirós A, Caraco Y, Williams-Diaz A, Brown ML, Du J, Pedley A, Assaid C, Strizki J, Grobler JA, Shamsuddin HH, Tipping R, Wan H, Paschke A, Butterton JR, Johnson MG, De Anda C. Molnupiravir for Oral Treatment of Covid-19 in Nonhospitalized Patients. N. Engl. J. Med. 2022, 386, 509–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Painter WP, Holman W, Bush JA, Almazedi F, Malik H, Eraut NC, Morin MJ, Szewczyk LJ, Painter GR. Human safety, tolerability, and pharmacokinetics of molnupiravir, a novel broad-spectrum oral antiviral agent with activity against SARS-CoV-2. Antimicrobial agents and chemotherapy, 2021, 65(5), 10–1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mahase E. Covid-19: Pfizer’s paxlovid is 89% effective in patients at risk of serious illness, company reports. British. Med. Journal, 2021, 375, n2713. [DOI] [PubMed] [Google Scholar]
- 19.Liu J, Pan X, Zhang S, Li M, Ma K, Fan C, Lv Y, Guan X, Yang Y, Ye X, Deng X, Wang Y, Qin L, Xia Z, Ge Z, Zhou Q, Zhang X, Ling Y, Qi T, Wen Z, Huang S, Zhang L, Wang T, Liu Y, Huang Y, Li W, Du H, Chen Y, Xu Y, Zhao Q, Zhao R, Annane D, Qu J, Chen D. Efficacy and safety of Paxlovid in severe adult patients with SARS-Cov-2 infection: a multicenter randomized controlled study. The Lancet. 2023, 33, 100694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wu F, Zhao S, Yu B, Chen Y-M, Wang W, Song Z-G, Hu Y, Tao Z-W, Tian J-H, Pei Y-Y, Yuan M-L, Zhang Y-L, Dai F-H, Liu Y, Wang Q-M, Zheng J-J, Xu L, Holmes EC, Zhang Y-Z. A New Coronavirus Associated with Human Respiratory Disease in China. Nature, 2020, 579 (7798), 265–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Baranov PV, Henderson CM, Anderson CB, Gesteland RF, Atkins JF, Howard MT. Programmed ribosomal frameshifting in decoding the SARS-CoV genome. Virology, 2005, 332, 498–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zhang L, Lin D, Sun X, Curth U, Drosten C, Sauerhering L, Becker S, Rox K, Hilgenfeld R. Crystal Structure of SARS-CoV-2 Main Protease Provides a Basis for Design of Improved α-Ketoamide Inhibitors. Science, 2020, 368 (6489), 409–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ziebuhr J, Snijder EJ, Gorbalenya AE. Virus-encoded proteinases and proteolytic processing in the Nidovirales. J Gen. Virol, 2000, 81, 853–879. [DOI] [PubMed] [Google Scholar]
- 24.Jin Z, Du X, Xu Y, Deng Y, Liu M, Zhao Y, Zhang B, Li X, Zhang L, Peng C, Duan Y, Yu J, Wang L, Yang K, Liu F, Jiang R, Yang X, You T, Liu X, Yang X, Bai F, Liu H, Liu X, Guddat LW, Xu W, Xiao G, Qin C, Shi Z, Jiang H, Rao Z, Yang H. Structure of Mpro from SARS-CoV-2 and Discovery of Its Inhibitors. Nature, 2020, 582 (7811), 289–293. [DOI] [PubMed] [Google Scholar]
- 25.Báez-Santos YM, St John SE, Mesecar AD. The SARS-coronavirus papain-like protease: structure, function and inhibition by designed antiviral compounds. Antiviral Res, 2015, 115, 21–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ghosh AK, Brindisi M, Shahabi D, Chapman ME, Mesecar AD. Drug development and medicinal chemistry efforts toward SARS-coronavirus and Covid-19 therapeutics. ChemMedChem, 2020, 15, 907–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ghosh AK, Mishevich JL, Mesecar AD, Mitsuya H. Recent Drug Development and Medicinal Chemistry Approaches for the Treatment of SARS-CoV-2 Infection and COVID-19. ChemMedChem, 2022, 17, e202200440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Alugubelli YR, Geng ZZ, Yang KS, Shaabani N, Khatua K, Ma XR, Vatansever EC, Cho CC, Ma Y, Xiao J, Blankenship LR. A systematic exploration of boceprevir-based main protease inhibitors as SARS-CoV-2 antivirals. Eur. J. Med. Chem, 2022, 240:114596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kneller DW, Li H, Phillips G, Weiss KL, Zhang Q, Arnould MA, Jonsson CB, Surendranathan S, Parvathareddy J, Blakeley MP, Coates L. Covalent narlaprevir-and boceprevir-derived hybrid inhibitors of SARS-CoV-2 main protease. Nat Commun, 2022, 13(1)2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Marzolini C, Kuritzkes DR, Marra F, Boyle A, Gibbons S, Flexner C, Pozniak A, Boffito M, Waters L, Burger D, Back DJ, Khoo S. Recommendations for the Management of Drug-Drug Interactions Between the COVID-19 Antiviral Nirmatrelvir/Ritonavir (Paxlovid) and Comedications. Clinical Pharmacology and Therapeutics, 2022, 112, 1191–1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Marzolini C, Kuritzkes DR, Marra F, Boyle A, Gibbons S, Flexner C, Pozniak A, Boffito M, Waters L, Burger D, Back D, Khoo S. Prescribing Nirmatrelvir-Ritonavir: How to Recognize and Manage Drug-Drug Interactions. Ann. Intern. Med, 2022, 175, 744–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ritonavir-Boosted Nirmatrelvir (Paxlovid). COVID-19 Treatment Guidelines. https://www.covid19treatmentguidelines.nih.gov/therapies/antivirals-including-antibody-products/ritonavir-boosted-nirmatrelvir--paxlovid-/ (accessed 2023–10-08).
- 33.Wang L, Berger NA, Davis PB, Kaelber DC, Volkow ND, Xu R. COVID-19 rebound after Paxlovid and Molnupiravir during January-June 2022. MedRxiv, 2022. [Google Scholar]
- 34.Greasley SE, Noell S, Plotnikova O, Ferre R, Liu W, Bolanos B, Fennell K, Nicki J, Craig T, Zhu Y, Stewar AE. Structural basis for the in vitro efficacy of nirmatrelvir against SARS-CoV-2 variants. J. Biol. Chem, 2022. 298(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ghosh AK, Yadav M. Synthesis of Optically Active SARS-CoV-2 Mpro Inhibitor Drug Nirmatrelvir (Paxlovid): An Approved Treatment of COVID-19. Org. Biomol. Chem, 2023, 21 (28), 5768–5774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bai B, Belovodskiy A, Hena M, Kandadai AS, Joyce MA, Saffran HA, Shields JA, Khan MB, Arutyunova E, Lu J, Bajwa SK, Hockman D, Fischer C, Lamer T, Vuong W, van Belkum MJ, Gu Z, Lin F, Du Y, Xu J, Rahim M, Young HS, Vederas JC, Tyrrell DL, Lemieux MJ, Nieman JA. Peptidomimetic α-Acyloxymethylketone Warheads with Six-Membered Lactam P1 Glutamine Mimic: SARS-CoV-2 3CL Protease Inhibition, Coronavirus Antiviral Activity, and in Vitro Biological Stability. J. Med. Chem, 2022, 65 (4), 2905–2925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhai Y, Zhao X, Cui Z, Wang M, Wang Y, Li L, Sun Q, Yang X, Zeng D, Liu Y, Sun Y, Cyanohydrin as an anchoring group for potent and selective inhibitors of enterovirus 71 3C protease. J. Med. Chem, 2015, 58, 9414–9420. [DOI] [PubMed] [Google Scholar]
- 38.Hanessian S, Margarita R. 1, 3-asymmetric induction in dianionic allylation reactions of amino acid derivatives-synthesis of functionally useful enantiopure glutamates, pipecolates and pyroglutamates. Tetrahedron letters, 1998, 39, 5887–5890. [Google Scholar]
- 39.Anson BJ, Chapman ME, Lendy EK, Pshenychnyi S, Richard TD, Satchell KJ, Mesecar AD. Broad-spectrum inhibition of coronavirus main and papain-like proteases by HCV drugs. Res. Square, 2020, preprint. DOI: 10.21203/rs.3.rs-26344/v1. [DOI] [Google Scholar]
- 40.Hattori SI, Higashi-Kuwata N, Hayashi H, Allu SR, Raghavaiah J, Bulut H, Das D, Anson BJ, Lendy EK, Takamatsu Y, Takamune N. A small molecule compound with an indole moiety inhibits the main protease of SARS-CoV-2 and blocks virus replication. Nature communications, 2021, 12, 668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tsuji K, Ishii T, Kobayakawa T, Higashi-Kuwata N, Shinohara K, Azuma C, Miura Y, Nakano H, Wada N, Hattori S, Bulut H, Mitsuya H, Tamamura H. Journal of Medicinal Chemistry, 2023, 66, 13516–13529. [DOI] [PubMed] [Google Scholar]
- 42.Maeda K; Higashi-Kuwata N; Kinoshita N; Kutsuna S; Tsuchiya K; Hattori S-I; Matsuda K; Takamatsu Y; Gatanaga H; Oka S; Sugiyama H; Ohmagari N; Mitsuya H Neutralization of SARS-CoV-2 with IgG from COVID-19-convalescent plasma. Sci. Rep, 2021, 11, 5563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Please see supplementary data section, Table 1.
- 44.Ghosh AK, Raghavaiah J, Shahabi D, Yadav M, Anson BJ, Lendy EK, Hattori SI, Higashi-Kuwata N, H..Mitsuya, A. D. Mesecar. Indole Chloropyridinyl Ester-Derived SARS-CoV-2 3CLpro Inhibitors: Enzyme Inhibition, Antiviral Efficacy, Structure-Activity Relationship, and X-ray Structural Studies. J Med Chem, 2021, 64, 19, 14702–14714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Otwinowski Z, Minor W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol, 1997, 276, 307–326. [DOI] [PubMed] [Google Scholar]
- 46.Adams PD, Afonine PV, Bunkóczi G, Chen VB, Davis IW, Echols N, Headd JJ, Hung LW, Kapral GJ, Grosse-Kunstleve RW, McCoy AJ, Moriarty NW, Oeffner R, Read RJ, Richardson DC, Richardson JS, Terwilliger TC, Zwart PH. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr D Biol Crystallogr, 2010, 66(Pt 2), 213–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Moriarty NW, Grosse-Kunstleve RW, Adams PD. electronic Ligand Builder and Optimization Workbench (eLBOW): a tool for ligand coordinate and restraint generation. Acta Crystallogr D Biol Crystallogr, 2009, 65(Pt 10), 1074–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Emsley P, Lohkamp B, Scott WG, Cowtan K. Features and development of Coot. Acta Crystallogr D Biol Crystallogr, 2010, 66(Pt 4), 486–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
X-ray crystallographic data for compound 5e has been deposited at the PDB (8UND). Spectral data, assay, and X-ray data supporting this article are available for public use.









