SARS-CoV-2 methyltransferase nsp10-16 in complex with natural and drug-like purine analogs for guiding structure-based drug discovery

  1. Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany;
  2. Department of Virology, Bernhard-Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359 Hamburg, Germany;
  3. Universität Hamburg, Center for Bioinformatics, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany
  4. The Hamburg Centre for Ultrafast Imaging, Luruper Chaussee 149, 22761 Hamburg, Germany;
  5. Institute of Pharmaceutical and Biomedical Science (IPBS), Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany;
  6. Department of Physics, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, public reviews, and a provisional response from the authors.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Yang Yang
    Iowa State University
  • Senior Editor
    Amy Andreotti
    Iowa State University, Ames, United States of America

Reviewer #1 (Public Review):

Summary:

This manuscript describes the crystallographic screening of a number of small molecules derived from the natural substrates S-adenosyl methionine (SAM) and adenine, against the SARS-CoV-2 2'-O-methyltransferase NSP16 in complex with its partner NSP10. High-quality structures for several of these are presented together with efforts to evaluate their potential biophysical binding and antiviral activities. The structures are of high quality and the data are well presented but do not yet show potency in biophysical binding. They only offer limited insights into the design of inhibitors of NSP16/10.

Strengths:

The main strengths of the study are the high quality of the structural data, and associated electron density maps making the structural data highly accurate and informative for future structure-based design. These results are clearly presented and communicated in the manuscript. Another strength is the authors' attempts to probe the binding of the identified fragments using biophysical assays. Although in general the outcome of these experiments shows negative data or very weak binding affinities the authors should be commended for attempting several techniques and showing the data clearly. This study is also useful as an example of the complexities associated with drug discovery on a bi-substrate target such as a methyltransferase, several of the observed binding poises were unexpected with compounds that are relatively similar to substrates binding in different parts of the active site or other unexpected orientations. This serves as an example of how experimental structural information is still of crucial importance to structure-based drug design. In general, the claims in the manuscript are well supported by the data.

Weaknesses:

The main limitations of the study are that the new structures generated in the study are fairly limited in terms of chemical space being similar to either SAM or RNA-CAP analogues. It feels a little bit of a lost opportunity to expand this to more diverse ligands which may reveal potential inhibitors that are distinct from current methyltransferase inhibitors based on SAM analogues and truly allow a selective targeting of this important target.

Another limitation is the potentially misleading nature of the antiviral assays. It is not possible to say if these compounds display on-target activity in these assays or even if the inhibition of NSP16/10 would have any effect in these assays. Whilst the authors do mention these points I think this should be emphasized more strongly.

Minor critical points:

The authors state that their crystals and protein preps have co-purified SAM occupying the active site of the crystals. Presumably, this complicates the interpretation of electron density maps as many of the ligands share overlap with the existing SAM density making traditional analysis of difference maps challenging. The authors did not utilize the PanDDA analysis for this step, perhaps this is related to the presence of SAM in the ground state datasets? Also, occupancies are reported in the manuscript in some cases to two significant figures, this seems to be an overestimation of the ability of refinement to determine occupancy based on density alone and the authors should clarify how these figures were reached.

The molecular docking approach to pre-selection of library compounds to soak did not appear to be successful. Could the authors make any observations about the compounds selected by docking or the docking approach used that may explain this?

Reviewer #2 (Public Review):

Summary:

The study by Kremling et al. describes a study of the nsp16-nsp10 methyl transferase from SARS CoV-2 protein which is aimed at identifying inhibitors by x-ray crystallography-based compound screening.
A set of 234 compounds were screened resulting in a set of adenosine-containing compounds or analogues thereof that bind in the SAM site of nsp16-nsp10. The compound selection was mainly based on similarity to SAM and docking of commercially available libraries. The resulting structures are of good quality and clearly show the binding mode of the compounds. It is not surprising to find that these compounds bind in the SAM pocket since they are structurally very similar to portions of SAM. Nevertheless, the result is novel and may be inspirational for the future design of inhibitors. Following up on the crystallographic screen the identified compounds were tested for antiviral activity and binding to np16-nsp10. In addition, an analysis of similar binding sites was presented.

Strengths:

The crystallography is solid and the structures are of good quality. The compound binding constitutes a novel finding.

Weaknesses:

The major weakness is the mismatch between antiviral activity and binding to the target protein. Only one of the compounds could be demonstrated to bind to the nsp16-nsp10 protein. By performing a displacement experiment using ITC Sangivamycin is concluded to bind with a Kd > 1mM. However, the same compound displays antiviral activity with an EC50 of 0.01 microM. Even though the authors do not make specific claims that the antiviral effect is due to inhibition of nsp16-nsp10, it is implicit. If the data is included, it should state specifically that the effect is not likely due to nsp16-nsp10 inhibition.

The structure of the paper and the language needs quite a lot of work to bring it to the expected quality.

Technical point:

Refinement of crystallographic occupancies to single digit percentage is not normally supported by electron density.

Author response:

eLife assessment

This important study describes the crystallographic screening of a number of small molecules against a viral enzyme critical for the 5' capping of SARS-CoV-2 RNA and viral replication. While the high-quality crystal structures and complementary biophysical assays in this study provide solid evidence to support the major claims regarding how these small molecule compounds bind to the viral enzyme, the mismatch between the antiviral activity and binding to the viral enzyme of several small molecule compounds could have been more thoroughly investigated or discussed. This paper would be of interest to the fields of coronavirus biology, structural biology, and drug discovery.

We do fully agree that the antiviral assay results could be brought better into context clarifying that the antiviral effects of tubercine and its derivates are due to off-target effects.

Reviewer #1 (Public Review):

Summary:

This manuscript describes the crystallographic screening of a number of small molecules derived from the natural substrates S-adenosyl methionine (SAM) and adenine, against the SARS-CoV-2 2'-O-methyltransferase NSP16 in complex with its partner NSP10. High-quality structures for several of these are presented together with efforts to evaluate their potential biophysical binding and antiviral activities. The structures are of high quality and the data are well presented but do not yet show potency in biophysical binding. They only offer limited insights into the design of inhibitors of NSP16/10.

Strengths:

The main strengths of the study are the high quality of the structural data, and associated electron density maps making the structural data highly accurate and informative for future structure-based design. These results are clearly presented and communicated in the manuscript. Another strength is the authors' attempts to probe the binding of the identified fragments using biophysical assays. Although in general the outcome of these experiments shows negative data or very weak binding affinities the authors should be commended for attempting several techniques and showing the data clearly. This study is also useful as an example of the complexities associated with drug discovery on a bi-substrate target such as a methyltransferase, several of the observed binding poises were unexpected with compounds that are relatively similar to substrates binding in different parts of the active site or other unexpected orientations. This serves as an example of how experimental structural information is still of crucial importance to structure-based drug design. In general, the claims in the manuscript are well supported by the data.

Weaknesses:

The main limitations of the study are that the new structures generated in the study are fairly limited in terms of chemical space being similar to either SAM or RNA-CAP analogues. It feels a little bit of a lost opportunity to expand this to more diverse ligands which may reveal potential inhibitors that are distinct from current methyltransferase inhibitors based on SAM analogues and truly allow a selective targeting of this important target.

It is true that it makes sense to screen for more diverse compounds to expand to a more diverse ligand set and we do hope our study motivates to do so. Given the limited number of crystal structures of nsp10-16 with potential drug molecules, the aim of this study was to upgrade the data base with new complex structures to have a pool of complex structures for future compound designs with increased selectivity. Furthermore, some of the hits are known inhibitors of similar enzymes and most prominent and potent methyltransferase inhibitors are structurally related to SAM, like sinefungin and tubercidine. We do think that knowing which SAM compounds or fragments of SAM are able to bind in the nsp10-16 active site is highly valuable for further specific and optimized inhibitor design.

Another limitation is the potentially misleading nature of the antiviral assays. It is not possible to say if these compounds display on-target activity in these assays or even if the inhibition of NSP16/10 would have any effect in these assays. Whilst the authors do mention these points I think this should be emphasized more strongly.

That is a very valid point and we do not believe that the antiviral activity is based on on-target effects. We do agree that the way it is currently presented can be considered misleading and we indeed clarify this point in the revised version.

Minor critical points:

The authors state that their crystals and protein preps have co-purified SAM occupying the active site of the crystals. Presumably, this complicates the interpretation of electron density maps as many of the ligands share overlap with the existing SAM density making traditional analysis of difference maps challenging. The authors did not utilize the PanDDA analysis for this step, perhaps this is related to the presence of SAM in the ground state datasets? Also, occupancies are reported in the manuscript in some cases to two significant figures, this seems to be an overestimation of the ability of refinement to determine occupancy based on density alone and the authors should clarify how these figures were reached.

We have used PanDDA in parallel for hit finding. We however did not see any advantages for this target over the hit finding results from the visual inspection. This is probably as mentioned because of SAM being present is the “ground state” which complicates the PanDDA map calculations.

Regarding the occupancies, we fully agree with this comment and change it to reasonable digits and clarify how the figures were reached.

The molecular docking approach to pre-selection of library compounds to soak did not appear to be successful. Could the authors make any observations about the compounds selected by docking or the docking approach used that may explain this?

Yes, it is a good point to give possible explanations why the docking approach was not successful to facilitate similar approaches in future studies.

Reviewer #2 (Public Review):

Summary:

The study by Kremling et al. describes a study of the nsp16-nsp10 methyl transferase from SARS CoV-2 protein which is aimed at identifying inhibitors by x-ray crystallography-based compound screening.
A set of 234 compounds were screened resulting in a set of adenosine-containing compounds or analogues thereof that bind in the SAM site of nsp16-nsp10. The compound selection was mainly based on similarity to SAM and docking of commercially available libraries. The resulting structures are of good quality and clearly show the binding mode of the compounds. It is not surprising to find that these compounds bind in the SAM pocket since they are structurally very similar to portions of SAM. Nevertheless, the result is novel and may be inspirational for the future design of inhibitors. Following up on the crystallographic screen the identified compounds were tested for antiviral activity and binding to np16-nsp10. In addition, an analysis of similar binding sites was presented.

Strengths:

The crystallography is solid and the structures are of good quality. The compound binding constitutes a novel finding.

Weaknesses:

The major weakness is the mismatch between antiviral activity and binding to the target protein. Only one of the compounds could be demonstrated to bind to the nsp16-nsp10 protein. By performing a displacement experiment using ITC Sangivamycin is concluded to bind with a Kd > 1mM. However, the same compound displays antiviral activity with an EC50 of 0.01 microM. Even though the authors do not make specific claims that the antiviral effect is due to inhibition of nsp16-nsp10, it is implicit. If the data is included, it should state specifically that the effect is not likely due to nsp16-nsp10 inhibition.

We do believe that the antiviral data are valuable and should be published within this work. We also agree with the comment that it should be clearly stated that the antiviral effect is not likely because of nsp10-16 inhibition and we will optimize that accordingly.

The structure of the paper and the language needs quite a lot of work to bring it to the expected quality.

We will go through the manuscript again and further improve the structure and language as much as possible

Technical point:

Refinement of crystallographic occupancies to single digit percentage is not normally supported by electron density.

We agree with that point and correct it in the revised version.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation