Preclinical characterisation
Determination of RSM01 epitope
Competition analysis by BLI with known anti-RSV monoclonal antibodies including MEDI8897* (non YTE version of nirsevimab, site Ø), motavizumab/palivizumab (site II), 101F/RB1 (site IV), and REGN222 (site V) indicated that RSM01 binds to antigenic site Ø [18]. RSM01 parental antibody ADI-15618 bound F-P2 (linear epitope aa 196–226) in a dose-dependent manner confirming site Ø specificity (Table 1). No binding to the linear peptides covering the other antigenic sites including site Ø F-P1 (aa 62–95) was observed. MEDI8897 bound neither to F-P2 or F-P1 indicating a distinct epitope from RSM01 (Table 1).
Table 1
Response units for RSV monoclonal antibodies binding to biotinylated peptides from antigenic site Ø
Antibody | Concentration (nM) | Biot-F-P2 | Biot-F-P1 |
Motavizumab (site II) | 100 | -0.0358 | 0.0917 |
REGN2222 (site V) | 100 | -0.0332 | -0.0194 |
D25 (site Ø) | 100 | -0.0388 | -0.0212 |
ADI-15618 (site Ø) | 100 | 1.6265 | -0.0186 |
REGN2222 (site V) | 5000 | -0.0271 | -0.0029 |
MEDI8897 (site Ø) | 5000 | 0.0035 | -0.0382 |
ADI-15618 (site Ø) | 5000 | 4.7153 | 0.0179 |
Binding of peptide was analysed by BLI. Two biotinylated peptides from antigenic site Ø were F-P2 (aa 62–95) and F-P1 (aa 196–226). Biot biotinylated; Biot biotinylated; BLI bio-layer interferometry; RSV Respiratory syncytial virus; aa amino acid |
X-ray crystallography was employed for fine epitope mapping of RSM01; the structure of the antibody-antigen complex (ADI-15618 Fab bound to RSV pre-F ecto A2 DS-Cav1 trimer) was solved at 3.9Å resolution (unpublished observations). Contact residues in the primary interface between RSM01 CDRs (complementary-determining regions) and F-protein are all located in site Ø with the majority in F-P2 region in agreement with binding and peptide mapping data. The main contact residues in RSM01 F protein interface were identified as: K68, N70, G71, T72, K201, Q202, L203, P205, I206, K209, Q210, S211, C212, S213, I214, S215, N216, T219. The primary interface was used to select clinical isolates with variations in these residues to assess potential resistance to RSM01 (Supplement Table 2).
In-vitro neutralisation activity
To determine the breadth of RSM01 neutralising activity, a panel of RSV-A and B viruses was assembled. The panel included common laboratory strains A2, A-Long, B9320, B1, B-Wash/18537, and 19 clinical strains isolated during the 2002–2017 RSV seasons. The clinical isolates were selected to cover all available sequence polymorphisms in antigenic site Ø (Supplement Table 2) allowing assessment of the impact of existing variations on antibody activity thereby increasing the probability to identify strains with lower susceptibility to RSM01.
RSM01 exhibited highly potent neutralising activity against all tested RSV strains (Fig. 1, Supplement Table 3). It was 2-3-fold more potent than nirsevimab and approximately 51-196-fold more potent than palivizumab for RSV-A strains (Supplement Table 4). For RSV-B, RSM01 was 1–10 fold more potent than nirsevimab and 3–90 fold more potent than palivizumab (Supplement Table 4). Potency was in the single ng/mL range (0.7–6.4) for all RSV-A subtypes and most of RSV-B subtypes (Fig. 1, Supplement Table 3). Interestingly, a RSV-B Wash/18537 isolate that was initially used at Sigmovir (Rockville, MD) for cotton rat studies (“RSV-B Wash-Military”), was found to be completely resistant to nirsevimab (Fig. 1, Table 3). This phenotype was confirmed in vitro and could be linked to the additional L203I sequence variation in the RSV-F-protein which was not present in the original RSV-B Wash/18537 isolate from ATCC (Supplement Table 2). Although L203I is located within the RSM01 epitope it did not abolish neutralising activity of RSM01 (EC50 = 21.5 ng/mL; Figure. 1, Supplement Table 3). All later cotton rat studies were performed with the original strain not harbouring this additional mutation, restoring the in vivo efficacy of nirsevimab.
In 2017, the rise of an RSV-B mutant strain was responsible for the failure of the REGN2222 binding to antigenic site V of RSV-F [20]. As expected, due to the different binding site, an RSV-B virus isolated during the 2017 season (17–000478) that was resistant to REGN2222 was fully susceptible to RSM01 (Supplement Table 3).
Epitope conservation
To understand the epitope conservation, an RSV sequence alignment was performed in 2018 using available RSV-A (n = 1632) and B (n = 668) sequences from GenBank. Since laboratory strains are heavily passaged and do not represent a consensus sequence, a first alignment of the GenBank sequences was done, and the consensus sequence of F protein was derived. Two clinical isolates represented this consensus sequence of the F protein: RSV-A strain 13-005275 and RSV-B strain 07–00043. Of note, both clinical isolates were fully neutralised by RSM01 (Fig. 1, Supplement Table 3).
Based on these initial results the RSM01 epitope was found to be highly conserved. Only 2.5% of the published RSV-A genomes and 12.15% of RSV-B strains showed sequence polymorphisms in the binding site of RSM01 (Supplement Table 5). For RSV-A, S213R was the most frequent variation and the only polymorphism that occurred with a relative frequency of > 1% (1.84%). Several strains (e.g. RSV-A-Long) carrying this mutation were included in the panel for in vitro testing and were confirmed to be neutralised by RSM01 (Fig. 1, Supplement Table 3).
A similar trend was observed for RSV-B with most variations occurring at very low frequency of < 1% (Supplement Table 5). Only one variant, Q209K, was found to be present in 7.49% of the available RSV-B genomes. This polymorphism was tested with a clinical isolate (RSV-B 13-013576) and RSM01 binding remained unaffected (EC50 = 1.6ng/ml) (Fig. 1, Supplement Table 3). The remaining variations accounted for only 0.15%-0.75% of the published sequences (Supplement Table 5), and 3 of these variants (Q202R, L203I, Q209R) were covered in the RSV-isolate panel tested in vitro (Supplement Tables 2 and 3).
A second sequence alignment of RSV F proteins was performed in 2024 (Supplement Table 6). The number of RSV sequences in GenBank substantially increased since 2018 to ~ 6,000 for each RSV subtype. To assess which amino acids were predominant, the most dominant animo acids were identified. Amino acids variation for RSV-A in antigenic site Ø remained very low with < 0.1% variation and no major differences from the 2018 assessment in the RSM01 primary interface (Supplement Table 6). For RSV-B, more variations were seen in the primary interface though the majority remained at low levels.
The most dominant amino acid polymorphisms were found in positions 206, 209 and 211. In the 2018 analysis, the variant I206M was a rare amino acid substitution reported only in 0.15% of RSV-B strains. In the 2024 analysis, M was the dominant amino acid at position 206 (68% of isolates) with I (now designated M206I) found only in 32% of strains (Supplement Table 6). Similarly, while Q209R was rare (0.3%) based on the original alignment in 2018, R now became the dominant residue (69% of isolates) according to the updated analysis; Q (R209Q) was found in only 31% (Supplement Table 6). Of note, RSV-B-09-017069, an RSV-B isolate harboring the Q209R mutation was included in the in vitro testing and was neutralized by RSM01 (EC50 4ng/ml (Supplement Table 3). Overall, we found that about 30% of all sequences analysed had variations in 206 or 209 positions (i.e. amino acids other than M206 or R209) and of these 48% had variations in both positions. The M206I and R209Q variations were co-present in most of these viruses.
Another polymorphism that increased in frequency was found to be in position 211 (S211N). While less than 0.5% of all available sequences showed variation in this position in 2018, this increased to 11.89% in the updated analysis (Supplement Table 6). Binding and neutralisation for this variant need to be confirmed in future studies.
Generation of MARMs
The use of monoclonal antibodies against viruses raises potential concern about the emergence of MARMs. Such escape variants have been described earlier for the benchmark monoclonal antobodies used in this study, palivizumab and nirsevimab [22–27]. A total of 11 experiments using 8 different RSV strains and antibody concentrations ranging from 60 ng/mL to 50 µg/mL were performed during the lead selection/characterisation phase of RSM01 [18]. No escape mutants emerged against RSM01 (or parental antibody ADI-15618), independent of the antibody concentration tested. However, MARMs were readily obtained with palivizumab (at 2.5 µg/mL and 50 µg/mL) and MEDI8897*/nirsevimab (at 60 ng/mL or 50 µg/mL). The mutations associated with the breakthrough viruses under palivizumab or nirsevimab/MEDI8897* were detected as early as passage 2 (P2). Observed mutations in the antibody binding sites are shown in the supplement (Supplement Table 7). Of note, passaging of the clinical isolate RSV-B 07 000431 (representing the consensus RSV-F sequence) in the presence of nirsevimab resulted in the rapid emergence of a resistant mutant carrying the L203I mutation in the F protein. This mutation was previously found in the nirsevimab resistant RSV-B-Wash/18537 strain, suggesting that L203I is indeed responsible for the resistance phenotype.
Prophylactic efficacy of RSM01 in cotton rat model
At 1 mg/kg, RSM01 reduced viral load in the lungs by 3.0 and 2.8 logs, nirsevimab reduced viral load by 3.1 and 1.6 logs and palivizumab by 1.0 and 1.1 logs in RSV-A and RSV-B infected animals, respectively (Fig. 2). At 1 mg/kg, RSM01 reduced nasal viral load by 3.0 and 2.4 logs while nirsevimab reduced viral load by 3.3 and 0.2 logs and palivizumab did not reduce viral titers in RSV-A and RSV-B infected animals, respectively (Supplement Table 8). Overall RSM01 was as potent as nirsevimab in reducing viral load in lungs or nasal tissue for RSV-A and more potent than nirsevimab for RSV-B. Compared with palivizumab, RSM01 was more potent in both compartments and with both RSV subtypes. The EC90 in cotton rats for nirsevimab was reported as 6.8 µg/mL [28, 29]. Based on efficacy analysis in cotton rat challenge model, it appears EC90 of RSM01 may to be comparable to nirsevimab.
Human FcγR binding
In-vitro binding studies demonstrated that RSM01 can bind to all human FcγRs, including Fcγ receptor I (CD64), IIA167His (CD32A167His), IIA167Arg (CD32A167Arg), IIB (CD32B), IIIA176Val (CD16A176Val), IIIA176Phe (CD16A176Phe), and IIIB (CD16B) (Supplement Table 9).
RSM01 Phase 1 Clinical Trial
Participants’ baseline characteristics and disposition
The trial period was 16 Nov 2021 to 07 Dec 2022 and all participants were followed through day 151. Of the 56 randomised participants, 48 received RSM01, 8 received placebo, and 53 completed the trial (Fig. 3). Two participants, both from the RSM01 3000 mg IV cohort, were lost to follow-up, and 1 participant, from the RSM01 600 mg IM cohort, discontinued from the trial due to a sponsor decision (participant unavailable for final PK assessment). Overall, demographics and baseline characteristics were comparable between the RSM01 and placebo groups (Table 2).
Table 2
Demographics and Baseline Characteristics
Characteristic | RSM01 Cohorts | Total Placebo N = 8 |
300 mg IV, N = 6 | 300 mg IM, N = 6 | 1000 mg, IV N = 6 | 3000 mg, IV N = 6 | 600 mg IM, N = 24 | Total RSM01 N = 48 |
Median age, years (range) | 28 (24–36) | 32 (19–45) | 32 (20–41) | 25 (20–45) | 33 (19–48) | 30 (19–48) | 30 (25–39) |
Sex, n (%) Male | 5 (83) | 3 (50) | 1 (17) | 4 (67) | 13 (54) | 26 (54) | 3 (38) |
Race, n (%) White | 4 (67) | 4 (67) | 3 (50) | 5 (83) | 16 (67) | 32 (67) | 5 (63) |
Black | 1 (17) | 2 (33) | 2 (33) | 0 | 6 (25) | 11 (23) | 3 (38) |
Native Hawaiian or Other Pacific Islander | 0 | 0 | 0 | 1 (16.7) | 0 | 1 (2.1) | 0 |
Multiple | 1 (17) | 0 | 1 (17) | 0 | 2 (8) | 4 (8) | 0 |
BMI, kg/m2 Mean (SD) | 23.3 (2.0) | 25.4 (3.0) | 24.8 (2.8) | 24.5 (4.5) | 24.7 (3.2) | 24.6 (3.1) | 22.9 (2.1) |
BMI body mass index; IM intramuscular; IV intravenous; SD standard deviation |
Safety
Overall, 12 (25%) participants in the RSM01 group and 2 (25%) in the placebo group, experienced unsolicited TEAEs (Table 3). COVID-19, headache, and nausea were the only unsolicited TEAEs in ≥ 2 participants in any of the RSM01 cohorts. There was no pattern in the AEs by route or dose of RSM01. Most AEs were mild or moderate in severity. One (2.1%) participant in the RSM01 3000 mg IV cohort experienced a severe AE of increased blood pressure which was not considered related to RSM01. The participant’s blood pressure increased from 135/90 mmHg predose to a maximum systolic blood pressure of 156 mmHg and a maximum diastolic blood pressure of 103 mmHg post dose. The event was resolved the next day.
Table 3
AEs category | RSM01 Cohort, n (%) | Total Placebo N = 8 |
300 mg IV N= 6 | 300 mg IM N = 6 | 1000 mg IV N = 6 | 3000 mg IV N = 6 | 600 mg IM N = 24 | Total RSM01 N = 48 |
Any TEAE | 1 (17) | 2 (33) | 3 (50) | 3 (50) | 3 (13) | 12 (25) | 2 (25) |
Treatment-related TEAEs | 0 | 1 (17) | 2 (33) | 0 | 1 (4) | 4 (8) | 0 |
Severe of life threatening TEAEs | 0 | 0 | 0 | 1 (17) | 0 | 1 (2) | 0 |
TEAEs leading to dose interruption/withdrawal (IV participants only) | 0 | NA | 1 (17) | 0 | NA | 1 (2) | 0 |
Any SAE | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Any AESI | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Unsolicited TEAEs in ≥ 2 Participants | | | | | | | |
COVID-19 | 0 | 0 | 0 | 1 (17) | 1 (4) | 2 (4) | 0 |
Headache | 0 | 0 | 2 (33) | 0 | 0 | 2 (4) | 1 (13) |
Nausea | 0 | 0 | 2 (33) | 0 | 0 | 2 (4) | 0 |
Deaths | 0 | 0 | 0 | 0 | 0 | 0 | 0 |
Events are sorted by frequency of TEAEs in the total RSM01 group. Participants with multiple adverse events within a MedDRA Preferred Term were counted only once. AESI adverse event of special interest; IM intramuscular; IV intravenous; NA not applicable; SAE serious adverse event; TEAE treatment emergent adverse event. |
Treatment-related unsolicited AEs were reported in 4 (8.3%) participants (Table 3). One participant in the 300 mg IM cohort had injection site pruritus and one in the 600mg IM cohort had pruritus. One participant in the 1000 mg IV cohort had nausea and one had nausea, infusion site pain, and headache. All treatment related AEs were mild to moderate in severity. The infusion site pain led to dose interruption that lasted for 11 minutes, after which the dosing was completed. No SAEs, AEs leading to discontinuation of trial intervention, or death, or AESIs were reported in the trial.
-
Solicited systemic and local AEs were reported more frequently with RSM01 than with placebo (Supplement Table 10). Five (10.4%) participants in the RSM01 group reported headache and 2 (4.2%) reported tiredness. None of the participants reported fever, joint pain, muscle pain, nausea, vomiting, or diarrhea within 7 days after RSM01 administration. All systemic solicited AEs were mild or moderate in severity and the majority had durations of 1 or 2 days. Of the 11 (45.8%) participants in the 600 mg IM cohort who received two injections in one thigh, 1 reported a mild solicited systemic AE of tiredness on day 1. In the placebo group, 1 (12.5%) participant reported a systemic solicited AE of fever (duration of 3 days; mild in severity).
Laboratory assessments, vital signs measurements and ECG parameters measured at baseline and throughout the trial were generally comparable between the RSM01 and placebo groups. There were no individual clinically significant abnormalities and no clinically meaningful difference in toxicity grade shifts between the RSM01 and placebo groups during the trial.
PK
PK parameters for RSM01 in serum are shown in Table 4. In the 300 mg and 600 mg IM cohorts, peak concentrations in serum (Tmax) were reached between approximately 6 days and 8 days post dose (Fig. 4). RSM01 concentration and AUC increased dose-proportionally following IV and IM administration (Supplement Fig. 2). RSM01 was eliminated gradually, in a monophasic manner following IM administration and in a biphasic manner following IV administration (Supplement Fig. 3). While comparing serum PK profiles, the IM route showed higher individual variability in the clearance (CL), than IV route. The observed between-participant variability was in line with the anticipated variability for a monoclonal antibody following IM administration.
Table 4
Summary of RSM01 serum PK in healthy adults from phase 1 trial
PK Parameter | 300 mg IV N = 6 | 300 mg IM N = 6 | 600 mg IM N = 24 | 1000 mg IV N = 6 | 3000 mg IV N = 6 |
Tmax [day] | 0.05 (0.05–0.05) | 5.97 (5.06–27.9) | 7.22 (6.9–29.1) | 0.24 (0.07–1.07) | 0.26 (0.09–0.73) |
Cmax or C0 [µg/mL] | 98.2 (15.4) | 39.8 (12.4) | 90.7 (21.6) | 314 (34.1) | 1050 (23.5) |
AUClast [day*µg/mL] | 4095 (43.4) | 2691(56.0) | 6958 (40.7) | 14833(26.6) | 450000 (23.9) |
All data are presented as geometric mean (geometric %CV) except for Tmax which are presented as median (min, max). AUClast area under the curve from dosing to last measurable concentration; C0 initial concentration; Cmax maximum concentration; CV Coefficient of variance; IM intramuscular; IV intravenous; PK Pharmacokinetics; Tmax time to maximum concentration. |
Population PK modeling and simulation
The PK analysis dataset comprised 420 samples, of which 372 samples from 48 participants contained measurable PK observations. Forty-eight serum observations, representing 11% of the total, fell below the lower limit of quantitation (BLQ) for the assay. These BLQ samples were considered missing data in the pharmacokinetics modeling analysis.
The PK of RSM01 following IV and IM administration was well characterised by a 2-compartment model with a zero-order absorption (for IM) and first-order elimination. The model estimated the CL, central volume of distribution (Vc), peripheral volume of distribution (Vp) and inter compartment clearance (Q) at 0.002 L/hr, 3.2 L/hr, 2.18 L/hr, and 0.029 L/hr, respectively. The model-derived half-life in adults after a 300 IM dose was 78 days (95% CI 31–190). The bioavailability of RSM01 was estimated to be approximately 80%. Differences in PK parameters based on body size were accounted for by using fixed-exponent allometric relationships. No covariates were selected in the covariate analysis.
The population PK model was used to extrapolate and simulate dynamics within virtual populations representative of infants in North America and Africa. Predicted PK parameters for the RSM01 50-mg intramuscular (IM) dose given to the North American and African infant populations are summarised in Table 5. The highest C150 and Cmax were observed for the 0 to < 3 months old infants for both the North American and African infant populations. Among the three infant age subgroups, the median C150 was 31 µg/mL and 52 µg/mL for 0 to < 3 months old, 21 µg/mL and 25 µg/mL for 3 to < 6 months old, and 16 µg/mL and 20 µg/mL for 6 to < 12 months old, for the North American and African infant populations, respectively. Similar trend was observed for Cmax for both the North American and African infant populations.
Table 5
Predicted PK parameters of RSM01 exposure in virtual North American and African infant populations following a 50 mg IM dose
| 0 to < 3 Months (N = 500) | 3 to < 6 Months (N = 500) | 6 to < 12 Months (N = 500) |
North American Infants |
Weight (kg) | 5.9 (4.7, 7.5) | 7.6 (5.9, 9.3) | 9.4 (7.1, 11) |
Cmax (µg/mL) | 83 (60, 120) | 65 (46, 94) | 52 (38, 74) |
C150 (µg/mL) | 31 (12, 53) | 21 (6.3, 40) | 16 (3.6, 30) |
AUC (mg•day/mL) | 6.8 (4.4, 10) | 5.1 (3.2, 7.9) | 4.1 (2.4, 6.0) |
Fraction of infants with C150 > 6.8 µg/mL (%) | 98.2 | 93.6 | 88.8 |
African Infants |
Weight (kg) | 3.6 (2.4, 5.1) | 5.6 (4.4, 6.9) | 7.3 (5.4, 9.2) |
Cmax (µg/mL) | 130 (83, 220) | 84 (60, 120) | 66 (48, 94) |
C150 (µg/mL) | 52 (23, 91) | 25 (8.1, 49) | 20 (5.5, 38) |
AUC (mg•day/mL) | 11 (6.5, 18) | 6.4 (3.9, 9.8) | 5.2 (3.0, 8.4) |
Fraction of infants with C150 > 6.8 µg /mL (%) | 99.6 | 96.0 | 92.8 |
Data are presented as median (5th percentile, 95th percentile). AUC area under the RSM01 concentration-time curve; C150 trough RSM01 concentration at day 150; Cmax maximum RSM01 concentration; N number of subjects with available information; PK pharmacokinetics |
The population PK model was used to predict the proportion of infants who would have RSM01 concentration levels above the EC90 threshold of 6.8 µg/mL 150 days post dose, to determine the potential rate of protection per RSV season (Table 4 and Fig. 5). According to the simulations, in African infants, following an RSM01 dose of 50 mg IM, 99.6%, 96.0%, and 92.8% of infants in the age groups of 0 to < 3 months, 3 to < 6 months, and 6 to < 12 months, respectively, were predicted to maintain RSM01 concentrations above the EC90 threshold of 6.8 µg/ml. For North American infants, following an RSM01 dose of 50 mg IM, 98.2%, 93.6%, and 88.8% of infants for the same respective age groups were predicted to maintain RSM01 concentrations above 6.8 µg/ml. These simulation results indicated that a single dose of RSM01 50 mg could potentially provide protection to infants (0 to < 12 months old) for the entire RSV season.
Immunogenicity
The baseline ADA-positive rate was 2/48 (4.2%) in RSM01 group. The two baseline ADA positive participants were in the RSM01 600 mg IM cohort and one remained ADA-positive after RSM01 administration. One baseline ADA-negative participant in the RSM01 1000 mg IV cohort was categorized as postbaseline ADA-positive, for a treatment-emergent ADA-positive rate of 1/48 (2.1%) in the RSM01 group. All participants in the placebo group were ADA‑negative at baseline and throughout the trial.
RSV Neutralising Activity of RSM01 in participants’ serum
RSV neutralising activity was measurable in all serum samples including prior to the first dose, with no samples BLQ. Analysis of baseline corrected RSV neutralising activity showed that neutralising activity correlated with the RSM01 dose (Fig. 6).