Lab-adapted RSV-A2 develops a Leucine to Isoleucine mutation at position 305 under polyclonal selective pressure at bottleneck titers.
To understand how RSV mutates in response to antibody pressure, we performed an in vitro resistance and evolution assay in the presence of a neutralizing polyclonal anti-RSV antibody, a non-nucleoside polymerase inhibitor that we have previously discovered 32,33, and a DMSO control. The virus was harvested every two days for 20 passages. Viral titer was measured by qRT-PCR and viral genomes were next-generation sequenced following significant drops in titer, which we refer to hereafter as bottlenecks (Fig. 1a and 1c). Bottlenecks were of interest to us because we sought to determine if the mutations at these positions lead to viral evasion of neutralizing antibody pressure and subsequently increased viral titer. The first bottleneck we identified under polyclonal anti-RSV antibody selective pressure was at passage 5 (Fig. 1a). Additional bottlenecks at passages 9 and 13 were observed but reliable reads could not be obtained by NGS past passage 9 and the virus was undetectable past passage 13 by RT-qPCR. Sequencing of the passage 5 viral population revealed the RSV-F L305I mutation as the only non-synonymous mutation occurring in the majority of sequencing reads (Fig. 1b, S. Table 1). Here, 42.03% of reads contained L (CTA) while 57.93% of reads contained the mutated I (ATA) (Fig. 1b, S. Table 1). An increase in the proportion of RSV-F L305I was subsequently associated with a rebound in viral titer (Fig. 1a and b). The next two most frequently occurring non-synonymous mutations were RSV-F N276S and RSV-F V152I, occurring at 36.92% and 31.82% respectively (S. Table 2). Interestingly, the RSV-F N276S has previously been associated with Palivizumab resistance, and modern RSV strains appear to be mutating away from the historic N at position 276 to an S 34. To the best of our knowledge, an RSV-F V152I mutation has not been reported in the context of viral resistance. However, upon examination of published RSV-F sequences, we determined that 98.7% of sequences contain isoleucine (I) at position 152, suggesting that a V may be characteristic of the early RSV-A2 genotype and that most modern circulating RSV strains have since mutated away from this. The RSV-F L305I mutation also occurred upon the treatment with our synthesized RSV polymerase inhibitor 33 (Fig. 1c). At 10 µM and 25µM, RSV-F L305I occurs at passage 8 (17.25%) and passage 6 (41.53%) respectively but is undetectable by passage 20 at both concentrations (data not shown).
Published and clinical RSV isolates show that amino acid position 305 is subtype specific.
Since RSV-F L305I was the only mutation to emerge in the majority of sequenced reads under selective anti-RSV pressure, we sought that it is crucial to understand its importance and relevance to viral evolution and the impact of this mutation. To determine the biological relevance of the RSV-F L305I mutation, we analyzed published RSV-F sequences from the Virus Pathogen Resource (ViPR) database and representative clinical samples from previously sequenced RSV-A and B clinical isolates from hospitalized patients in Alberta, Canada, and Nationwide Children’s Hospital in Columbus, Ohio, during the 2014 to 2016 seasons 9. The percent identities between clinical isolates were determined (Fig. 1d). We found that RSV-F was one of the most conserved genes among our clinical isolates, in agreement with previous reports 26. RSV-F sequences shared greater than 99% identity within each individual subtype and 90.4% identity between RSV-A and RSV-B isolates (S. Table 3). Upon further analysis of our sequencing library of RSV clinical isolates 9, we noted that at position 305 almost all RSV-A and RSV-B sequences had a conserved L and I, respectively.
To support our modest sample size, we downloaded 3747 complete RSV-F sequences from the ViRP database and analyzed their amino acid composition (Fig. 1e and S. Table 4). As we observed in our clinical isolates, RSV-A and RSV-B subtypes differed in their amino acid composition at position 305, as others have previously noted 35. In addition to position 305, there were approximately 50 other residues throughout RSV-F that appeared to be subtype-specific (Fig. 1e). It is worth noting that no other amino acid, other than L or I, was found at position 305 in any RSV isolates, suggesting that the RSV-F L305I mutation is not simply an artifact of our in vitro evolution assay and may indeed serve a biological function.
An L305I mutation in the RSV-A2 fusion glycoprotein alters the conformation of the RSV-F protomer.
The constitutional isomers, leucine and isoleucine, inherently share several biochemical properties and differ structurally only in the position of a side chain methyl group (Fig. 2a). We asked whether this structural difference could have a steric effect on nearby amino acids in the RSV-F protein. To investigate this, we introduced the L305I mutation into the cryo-EM structure of the RSV-A2 prefusion F glycoprotein using the Schrödinger Small Molecule Discovery Suite followed by a molecular dynamics simulation of both proteins. Comparison of the wild-type RSV-F L305 and mutant RSV-F I305 models revealed that an L305I mutation induces a subtle protein-wide conformational change (Fig. 2b). Superimposition of wild-type and mutant RSV-F suggests that this structural change extends to a variety of binding sites including antigenic sites ∅, II, and IV (Fig. 2b and c).
RSV-F exists on the surface of the RSV virion as a dynamic trimer that has been shown to “breathe”, a phenomenon that can alter the accessibility of specific epitopes on trimerized RSV-F 36. To determine whether L305I affects trimer breathing, we evaluated the dynamics of RSV-F L305 and RSV-F I305 using Molecular Dynamics (MD) simulations (Fig. S1). Analysis of the MD trajectories revealed that the Cα of both systems each equilibrated around 200 ns of the simulation time with an average RMSD of 2.3 Å and 2.8 Å for RSV-F L305 and RSV-F I305 respectively, suggesting that the mutation has little effect on trimer breathing (Fig. S1a). Next, we investigated the flexibility of the system residues using atomic fluctuations expressed as a function of B-factor of the protein backbone atoms for both systems (Fig. S1b). The majority of protein residues were stable during the entire simulation with little flexibility observed in the region 250–305 for RSV-F I305 compared to that of the RSV-F L305 and with increased flexibility in the 420–500 region of RSV-F L305 compared to RSV-F I305 (Fig. S1b and S movie 1). The flexibility of the latter region could be attributed to the presence of a flexible loop that connects F1 and F2 protomers and mutations in this region have been previously utilized to develop a stable prefusion RSV F vaccine 37; however, the stabilization of the movement in this region in the L305I mutant could be attributed to effect of the mutation at position 305 and the effect has been distally extended to affect the overall dynamics and stability of the RSV F trimer. In summary, using computational simulations, we predict that there are flexibility differences between RSV-F L305 and RSV-F I305 proteins in certain regions, which could explain the observed structural alterations between the two.
To further validate the significance of the RSV-F L305I mutation, we compared the structure of the RSV-A2 pre-fusion F protein containing an asparagine or serine at position 228. We chose this position because analysis of published NCBI data highlighted it as another subtype-specific residue in RSV-F, that is Asn (N) in RSV-A and Ser (S) in RSV-B and is found distal to the binding site ∅ (Fig. 1e). Clustering of the MD trajectories suggested that the RSV-F S228 mutant did not show a noticeable conformational shift from the wild-type RSV-F N228. Atomic alignment of the RSV-F S228 and RSV-F N228 structures had an RMSD of 2.4 Å. The RMSD of both systems showed a stable structure during the whole simulation with an average RMSD of 2.9 Å and 2.7Å for the RSV-F N228 and RSV-F S228, respectively (Fig S1c). We also analyzed the atomic fluctuation to understand the regional flexibility of the structures. Our results indicate a similar trend in region flexibility between RSV-F N228 and RSV-F S228 suggesting that this mutation causes no to minimal conformational changes (Fig S1d). Taken together, our computational modeling predicts that introducing the conservative RSV-F L305I mutation into RSV-A2 can alter the structural confirmation of the entire RSV-F glycoprotein and that this observed difference is likely not due to alterations in trimer breathing.
A conservative Leucine to Isoleucine mutation at position 305 in an RSV reverse genetics model reduces infectivity of viral particles and susceptibility to human sera.
Given that our models predict a structural difference in RSV-A2 fusion proteins containing either an L or an I at position 305, we wondered whether this single mutation would have a functional impact on RSV in vitro. To test this, we used a recombinant reverse genetics model of RSV-A2 based on the RW30 backbone (rgRSV) 38,39. We added in an L305I mutation via Gibson assembly and the resulting plasmid was sequenced to confirm the presence of RSV-F I305. First, we looked at whether there were any changes in growth characteristics between the wild-type rgRSV-A2 L305 and mutant rgRSV-A2 I305, which we will refer to as RSV WT and RSV L305I respectively. Since RSV-B isolates have been found to grow slower than RSV-A isolates in tissue culture, we hypothesized that RSV L305I might also grow slower than RSV WT 40,41. To test this, we created a one-step growth curve of RSV WT and RSV L305I (Fig. 2d) in which HeLa cells were infected with equivalent MOIs, and the media was collected every 5 hrs over 100 hrs. Subsequent viral particle release was measured by RT-qPCR. We found no significant difference in the number of viral transcripts released between RSV WT and RSV L305I (Fig. 2d). However, when we measured foci of infection, we found that RSV L305I resulted in significantly fewer foci than RSV WT (Fig. 2e). This suggests that although similar amounts of transcripts are created by the two viruses, the virions produced by RSV L305I appear to be significantly less infectious (Fig. 2e).
Taking into account our modeling data, we asked whether the sensitivity of RSV to human sera may be altered by the L305I mutation. We obtained polyclonal sera from otherwise healthy full-term infants that had confirmed infections with RSV-A or RSV-B. We decided to use infant sera to reduce confounding results from older patients which may be complicated by immune memory to previous RSV infections. However, our sera samples were obtained from infants ranging in age from 2 weeks − 13.3 months. Although maternal RSV antibodies have been previously reported to be mostly absent by 3 months of age, we cannot rule out whether these infants had received passive RSV antibodies from their mothers, particularly in sera samples from infants less than 3 months of age 42. Briefly, HeLa cells were infected at an MOI of 0.3 with RSV WT or RSV L305I incubated with a 10− 3 dilution of each sera sample for 1 hr and analyzed by flow cytometry (Fig. 3). As was expected, different sera samples had varying levels of neutralization to RSV WT and RSV L305I (Fig. 3a). Though we found that overall RSV L305I was significantly more resistant to human sera than RSV WT (Fig. 3b). This is in line with what we observed in our in vitro evolution experiment, wherein the introduction of the L305I mutation led to a temporary rebound in viral titer (Fig. 1a).
Computational modeling of the RSV-F L305I mutation shows a structural shift in the RSV-F antigenic site II.
Currently, prophylactic monoclonal antibodies are the therapeutic of choice to prevent RSV infection in infants. PZMB and Nirsevimab have been FDA-approved for use in infants and a third antibody - Clesrovimab - is in late-phase clinical trials 4. These antibodies target antigenic sites II, ∅, and IV respectively, on RSV-F (Fig. 4a). We sought to investigate the impact that the L305I mutation may have in the context of these prophylactic treatments. Our in silico structural modeling shows that position 305 is located in antigenic site III, typically considered a weak antigenic site (Fig. 2b). However, we also observed that position 305 is located directly behind PZMB binding site II 25 in RSV-F (Fig. 2a and 4b). Using molecular dynamics simulations at over 500 ns, we found that the L305I mutation led to a conformational change in antigenic site II (Fig. 4b). Superimposition of site II RSV-F L305 and RSV-F I305 revealed not only a movement in the PZMB binding site but also a change in the orientation of several residues in which the L305I mutation forced residue N254 to change orientation outwards by 137.4˚ (Fig. 4c). In addition, Q279 has a bond angle change of 137.2˚ from the wildtype RSV-F L305 to the mutant RSV-F I305 (Fig. 4c). In summary, it appears that the steric effects caused by the L305I mutation extend to and affect the structural confirmation of antigenic site II.
The structural change elicited by RSV-F L305I alters antibody binding affinity to site II and viral susceptibility to palivizumab.
To determine whether the predicted change in the structure of binding site II affects PZMB neutralization, we conducted an in vitro PZMB neutralization assay with RSV WT and RSV L305I. We found that RSV WT was more susceptible to palivizumab (EC50 = 91.26 ng/mL) than RSV L305I (EC50 = 244.1 ng/mL) (Fig. 4d). Even at a maximum concentration of 105 ng/mL, PZMB was unable to fully neutralize RSV L305I, whereas RSV WT appeared to be almost fully neutralized by 103 ng/mL of PZMB (Fig. 4d). To further validate the change in PZMB sensitivity to neutralization, we directly assessed the binding affinity of PZMB to RSV-F using surface plasmon resonance (SPR). To do this we used a recombinant pre-fusion stabilized RSV-F protein trimer called DS-Cav118. Wildtype (DS-Cav1 L305) and mutant (DS-Cav1 I305) constructs were used to measure the kinetics of bivalent IgG PZMB binding to protein. We observed a slight, but not significant, decrease in the KD value, for the mutant DS-Cav1 I305 (KD = 0.10 nM) as compared with the WT DS-Cav1 L305 (KD = 0.29 nM) (Fig. 4e and f, S. Table 6). Since KD is inversely proportional to binding affinity, this suggests that there is no observable change to PZMB binding affinity induced by the L305I mutation.
RSV-F L305I alters the structure of RSV-F antigenic sites Ø and IV, and subsequent binding affinity of monoclonal antibodies.
Following our characterization of the effect of L305I on binding site II, we asked whether the same mutation could similarly affect more distant antigenic sites. Several therapeutic monoclonal antibodies that target various RSV-F antigenic sites are of particular interest to us, including the recently approved Nirsevimab and the phase III clinical trial antibody Clesrovimab, which target binding sites Ø and IV respectively 4,43. To study these binding sites, we used monoclonal antibodies D25 and 101F. D25 is the parental antibody to Nirsevimab 44 while 101F and Clesrovimab both target binding site IV, specifically the epitopes spanning residues 427–438 and 426–447 respectively 45,46. Analysis of MD trajectories revealed that antigenic site Ø, at the apex of the RSV-F I305 monomer and trimer, was shifted by 9.1 Å for α1 and 10 Å for α5 helices when compared to RSV-F L305 (Fig. 5a), while site IV shifted by about 8 Å (Fig. 5b). Interestingly, these results suggest that the L305I mutation, which is located in antigenic site III, also affects the structural conformation of antigenic sites Ø and IV in RSV-F.
To confirm whether these structural changes have biological relevance, we evaluated the neutralizing potential of D25 and 101F to RSV WT and RSV L305I. We saw similar trends with monoclonal antibodies D25 and 101F as we did with PZMB. The EC50 value for D25 was higher against RSV L305I (19.53 ng/mL) than RSV WT (6.46 ng/mL) suggesting that the L305I mutation acts as a resistance mutation (Fig. 5c). Similar to our PZMB results, RSV L305I could not be fully neutralized even at a maximum antibody concentration of 104 ng/mL whereas RSV WT was fully neutralized by 102 ng/mL of antibody (Fig. 5c). Similarly, we observed that the EC50 value of 101F was also higher against RSV L305I (153.7 ng/mL) compared to RSV WT (71.27 ng/mL), suggesting that the L305I mutation also makes RSV less susceptible to 101F (Fig. 5d). To validate these findings, we again compared the binding kinetics of D25 and 101F to DS-Cav1 L305 and DS-Cav1 I305 RSV-F proteins using SPR. Consistent with our D25 neutralizations, we report an increased KD for DS-Cav1 I305 (4.40 nM) compared to DS-Cav1 L305 (0.84 nM), suggesting that the binding affinity of D25 is higher to the wildtype RSV-F than the mutant (Fig. 5e and g). SPR with 101F revealed similar results to PZMB, wherein the KD value for DS-Cav1 L305 (0.52 nM) and DS-Cav1 I305 (0.31 nM) was about the same, suggesting that the L305I mutation does not affect the binding affinity of 101F (Fig. 5f and h). This data suggests that the increased resistance to D25 can be attributed to a change in binding affinity towards RSV-F, where D25 exhibited a weaker binding to RSV-F L305I (Fig. e and g). However, it is not clear what the source of the increased resistance to PZMB and 101F is if the L305I mutation does not alter the binding affinity of these antibodies to prefusion RSV-F (Fig. 4 and Fig. 5). Given that antigenic sites II and IV are retained during the conversion of prefusion to post-fusion RSV-F, whereas site Ø is not, it is possible that the L305I mutation also has an effect on antibody binding to post-fusion RSV-F in these cases. Overall, our results suggest that the L305I mutation can elicit significant changes across at least RSV-F antigenic sites Ø, II, and IV, and subsequently impact the effectiveness of monoclonal antibodies that target these regions.