The Durability and Avidity of MPXV-specific Antibodies Induced by the Two-dose MVA-BN Mpox Vaccine ================================================================================================== * Aaron L. Oom * Angelica C. Kottkamp * Kesi K. Wilson * Miilani Yonatan * Stephanie Rettig * Heekoung Allison Youn * Michael Tuen * Yusra Shah * Ashley L. DuMont * Marie I. Samanovic * Ralf Duerr * Mark J. Mulligan * the NYC OSMI Study Group ## Abstract The 2022 global outbreak of clade IIb mpox was the first major outbreak of mpox outside of African nations. To control the outbreak, vaccination campaigns were begun using the third-generation orthopoxvirus vaccine MVA-BN. During the vaccination campaign, we launched the New York City Observational Study of Mpox Immunity (NYC OSMI). NYC OSMI is a longitudinal study that enrolled 171 MVA-BN vaccinees with or without prior smallpox vaccination and mpox convalescent individuals. Study participants had blood drawn prior to vaccination, after one dose, and after two doses. Mpox virus (MPXV) neutralizing titers in sera reach a comparable peak in naïve and experienced vaccinees. However, neutralizing titers return to baseline in less than one year for naïve individuals, while remaining elevated in those with prior smallpox vaccination. Both naïve and experienced individuals generate robust, immunodominant IgG responses against MPXV H3 and A35, but with significantly lower avidity in naïve vaccinees. Their vaccinia virus homologs H3 and A33 have previously been shown to be protective targets for orthopoxvirus infection and disease in mouse models. These data highlight a low avidity antibody response elicited by MVA-BN that is short-lived in naïve vaccinees. This study supports the need for studies of long-term protection from MVA-BN, the potential need for booster doses, and further development of next-generation orthopoxvirus vaccines. ## Introduction The 2022 global outbreak of clade IIb mpox (formerly known as monkeypox) was the first major outbreak of mpox outside of African nations with over 93,000 cases reported to date primarily in Europe and the Americas1. Over 31,000 cases have occurred in the US alone2. With cases primarily clustered among men who have sex with men, it was suggested that sexual transmission was playing a larger role in this outbreak3 compared to the routes of transmission typically noted in endemic cases4. To stem rising case numbers around the world, public health agencies launched awareness campaigns and began offering the MVA-BN two-dose vaccine series (Bavarian Nordic) in numerous clinics. MVA-BN (also known as IMVAMUNE, IMVANEX, or JYNNEOS) is a modified vaccinia Ankara vaccine that is FDA-approved to be administered twice subcutaneously (SC) with a 28-day interval between doses. It is a third-generation orthopoxvirus vaccine that is non-replicating as compared to the replication-competent, vaccinia virus (VACV)-based first-(e.g., DryVax) and second-generation (e.g., ACAM2000) vaccines. During the 2022 outbreak, limitations in the number of available doses led to an emergency use authorization (EUA) for a dose-sparing measure that used one-fifth the dose and was delivered intradermally (ID) rather than SC5. This change in route of administration was based on previous findings that this measure yielded comparable serological responses as the approved vaccination strategy6. Additionally, some public health agencies delayed second doses to maximize the number of individuals with at least a single dose, leading to variability in the timing of dose two. To examine the effects of these changes, we designed and conducted the New York City Observational Study of Mpox Immunity (NYC OSMI or OSMI). OSMI is a longitudinal study of MVA-BN vaccinees in NYC that began enrollment in November 2022. The present study includes additional samples from the New York University Langone Vaccine Center Biorepository to give a study range from July 2022, when vaccination began in NYC, to October 2023. We previously reported an interim analysis of OSMI where we showed a marked difference in antibody responses between MVA-BN vaccinees that had no prior smallpox vaccination (naïve vaccinees) and those that did (experienced vaccinees)7. That report highlighted the limited durability of MVA-BN serological responses in naïve recipients based on anti-mpox virus (MPXV) H3 antibody titers out to five months post dose two. We now report here an expanded dataset up to one-year post dose two using neutralization titers and a multiplexed immunoassay to measure binding antibodies and their avidity for a panel of MPXV proteins. While a number of studies have established the decades of robust immunity generated by VACV vaccination8–12, prior studies of MVA-BN durability have yielded more mixed results. Ilchman et al. found that VACV neutralizing titers wane to baseline levels within two years13, while Priyamvada et al. reported higher than baseline MPXV and VACV neutralizing titers at two years post-vaccination14. However, the latter study was conducted in the Democratic Republic of Congo where mpox is endemic, which may have impacted participant immunity. These long-term studies are also limited in their analyses and do not examine antibody responses against specific proteins or the avidity of those antibodies. Prior studies of the antibody repertoire following MVA-BN vaccination only investigated responses out to two or three months after vaccination15,16. We show here for the first time that the binding antibody titers generated by MVA-BN are low avidity after the two-dose primary series and show little to no avidity maturation by one-year post dose two. These data highlight a nondurable, low avidity response from the two-dose regimen of MVA-BN in naïve people that will likely require booster doses13,17. Of note, the CDC currently does recommend a booster dose of MVA-BN every two years for workers with ongoing occupational risk of exposure to the more virulent orthopoxviruses including MPXV18. ## Methods ### Study Design NYC OSMI investigates the immune response in adults with and without HIV who have received the MVA-BN vaccination or have recovered from mpox disease. The study is registered on [ClinicalTrials.gov](http://ClinicalTrials.gov) (NCT Number: [NCT05654883](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT05654883&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom)). OSMI includes blood draws for serum and peripheral blood monocytes at specified intervals, as illustrated in Figure 1. Participants had the opportunity to enroll either pre- or post-mpox vaccination, with the enrollment window extending up to 365 days post-first dose or mpox symptom onset. One hundred and seventy-one (171) adult participants have given informed consent to be part of the study, as approved by the NYU Grossman School of Medicine Institutional Review Board (protocol 22-01338). Participants were recruited from city vaccination centers as well as by word of mouth. In addition to demographic and clinical information, the study includes blood draws from before vaccination, after a single dose, and multiple study visits after the second dose. In this observational study, the cohort has a range of vaccination route combinations, dosing intervals, and demographics. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/01/29/2024.01.28.24301893/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2024/01/29/2024.01.28.24301893/F1) Figure 1: The New York City Observational Study of Mpox Immunity (OSMI) Diagram of study visits. The windows of time denoted for each visit are used throughout the paper in analyses of specific visits. Diagram generated in BioRender. Information regarding prior smallpox vaccination was gathered from the participants and confirmed through an adjudication process. For this process, the following factors were considered: physical examination of the upper arms for presence of scar, prior Bacillus Calmette-Guerin (BCG) vaccination, prior military service or other occupational risk requiring smallpox vaccination (i.e., including laboratory work), country of birth, year of migration to the U.S., and whether the year of vaccination was consistent with active smallpox vaccine distribution in the participants country of origin. A literature review was performed to determine when individual countries ended their routine smallpox vaccination campaigns. Review of smallpox vaccination status was conducted by two infectious diseases physicians. For people with HIV (PWH), CD4+ T cell counts were sourced from electronic medical records at the time of enrollment. The date and route of mpox vaccination were verified using electronic medical records when available or through participant’s description of the vaccination procedure. Additional control samples and pre-OSMI enrollment samples were pulled from the New York University Langone Vaccine Center Biorepository. ### MPXV virus stock propagation All mpox virus work was conducted in a certified ABSL3 facility. MPXV clade IIb, lineage B.1, was obtained from BEI Resources (NR-58622); this virus was isolated in Massachusetts, USA during the 2022 outbreak. Working stocks of MPXV were generated by plaque purification in Vero E6 cells (ATCC #CRL-1586) followed by 3 passages of propagation in Vero E6 cells at 37°C and a multiplicity of infection (MOI) of 0.01 for 3 days each. Virus was harvested by freeze-thaw lysing scraped cells followed by clarification by centrifugation to remove cell debris. Stocks were purified at each passage over 36% sucrose in TNE buffer (Quality Biological #351-302-101) at 32,900xg for 80 min at 4°C. Purified MPXV stocks were sequence-verified. ### Immunofluorescence-based MPXV microneutralization assay On the day before MPXV infection, 1.5×104 Vero E6 cells were plated in each well of a black 96-well assay plate in cell culture media (Dulbecco’s Modified Eagle Medium [DMEM] supplemented with penicillin/streptomycin, 2 mM L-glutamine, and 10% heat-inactivated fetal bovine serum [FBS]) and incubated at 37°C and 5% CO2. Participant serum was 2-fold serially diluted in infection media (DMEM lacking sodium pyruvate supplemented with 2% heat-inactivated FBS) starting at a 2.5-fold dilution (final starting dilution is 5-fold following addition of an equal volume of virus) for 8-points. Prior to infection, serum dilutions were incubated with MPXV (sufficient for MOI ≈ 0.01) for 1 hr at 37°C. Cells were washed with 1x phosphate-buffered saline (PBS) then incubated with serum-virus mixtures for 42 hrs at 37°C and 5% CO2. Following infection, plates were submerged in 10% formalin (Fisher Scientific #SF984) for 1 hr at RT. Fixed samples were rinsed with water then permeabilized and blocked with 3% bovine serum albumin (BSA) and 0.1% Triton X-100 in PBS for 30 min at RT. Samples were then incubated with a polyclonal rabbit anti-vaccinia virus Lister strain antibody (Abbexa #abx023200) diluted 1:1,000 in 3% BSA in PBS (blocking buffer) for 1 hr at RT. All plates were washed four times with PBS then stained with 1:2,000 dilution of donkey anti-rabbit AlexaFluor647 secondary (Thermo #A-31573) and 1.25 µg/mL DAPI in blocking buffer. Following staining, cells were washed four times with 1x PBS with 100 µL of PBS left in each well for imaging and quantification using the BioTek Cytation 7 Cell Imaging Multi-Mode Reader and Gen5 Image Prime software. MPXV neutralizing titers (ID50 values) were calculated from percent inhibition using GraphPad Prism 10.0.03 non-linear regression (variable slope with four parameters) with top and bottom constraints (100 and 0, respectively). ### Multiplexed immunoassay for binding antibodies and avidity Binding and avidity of MPXV-specific IgG in serum were measured using the Luminex platform for a 12-plex assay. Eight different MPXV proteins were included: A29 (Sino Biological #40891-V08E), A30 (Cell Sciences #YVV15001A), A35 (Sino Biological #40886-V08H), B16 (Cell Sciences #YVV17401A), B21 (Cell Sciences #YVV16301A), E8 (Cell Sciences #YVV13201A), H3 (Sino Biological #40893-V08H1), and L1 (Sino Biological #40889-V07E). In addition, quality control beads were included: rubella virus (RUBV) E1/E2 (ACROBiosystems #GL2-R5583), BSA (Thermo Scientific #J65097.22), human recombinant IgG1 (Sigma-Aldrich #I5154), and IC45 (Luminex #MRP1-045-01). Differing bead regions were coupled to antigen using the xMAP Antibody Coupling Kit (Luminex #40-50016) at the following concentrations: 10 pmol/1×106 beads for H3, A35, RUBV E1/E2, and human IgG1; 50 pmol/1×106 beads for E8 and A29; and 100 pmol/1×106 beads for A30, B21, L1, and BSA following prior optimization. Beads were multiplexed with 1,000 beads per region for each sample. All MPXV antigens are His-tagged and coupling was confirmed using an anti-His antibody (Abcam #ab27025). Samples were analyzed by first incubating equal volumes of complement-inactivated serum (heated for 30 min at 56°C) and multiplexed beads for 1 hr in the dark on a shaker at RT. All sera were analyzed at final dilutions of 1:100 and 1:2,000, prepared in technical duplicate in 1x PBS (-Ca2+ -Mg2+) with 0.01% BSA and 0.02% Tween-20 (PBS-TB). The beads were then washed twice with PBS-TB before being incubated with either PBS (-Ca2+ -Mg2+) or the chaotropic agent, 2M ammonium thiocyanate (NH4SCN) for 30 min in the dark on a shaker at RT; one technical replicate of each dilution was used for each condition. Beads were again washed and then incubated for 30 min as before with rabbit anti-human IgG H&L detection antibody (Abcam #ab97158) diluted to 2 µg/mL in PBS-TB. Finally, the beads were washed before a final incubation with streptavidin-PE (BioLegend #405204) diluted to 4 µg/mL in PBS-TB for 30 min as before. The beads were washed a final time and then run on a Luminex 200 instrument with sample volume set to 50 µL. Instrument settings were as follows: 50 bead minimum per region and DD gate set 6,000 to 17,000. Binding titers were calculated by measuring the area under the curve (AUC) of the median fluorescence intensity of the two dilutions that received PBS, not NH4SCN. To calculate avidity, the binding AUC from the NH4SCN condition was divided by the AUC from the PBS condition to determine a ratio that is reported as the avidity index. Samples were normalized by the inclusion of a positive control pool in each batch generated from pre-2022 controls with prior smallpox vaccination. ### Anti-MPXV H3 Enzyme-Linked Immunosorbent Assay (ELISA) Binding antibodies against MPXV H3 protein were measured via direct ELISA of participant serum as previously described7. Briefly, ninety-six well plates were coated with 0.5 µg/mL MPXV H3 protein (Sino Biological Inc., 40893-V08H1) diluted in PBS. Plates were blocked with PBS +0.05% Tween 20 (Thermo Fisher Scientific) (PBS-T) +5% non-fat milk (blocking buffer) for 1 hr at RT. Heat-inactivated serum samples were serially diluted in blocking buffer and incubated in wells for 2 hr at RT. Horseradish peroxidase (HRP)-conjugated goat-anti-human IgG (Southern BioTech, 2040-05) was diluted in blocking buffer (1:2,000) and added to each well to incubate for 1 hr at RT. Finally, plates were developed for 5 min with 3,3’,5,5’-Tetramethylbenzidine Peroxidase Substrate (Thermo Fisher Scientific) followed by 1N hydrochloric acid to halt the assay. Absorbance at 450 nm was measured on a Synergy 4 (BioTek) plate reader. Endpoint titers were determined for each sample by taking the reciprocal of the highest dilution that gives a positive reaction. Reaction positivity was determined by calculating 2 times the standard deviation plus the mean of 16 pre-2022 control samples with no prior history of smallpox vaccination. Endpoint titers were calculated by interpolating the cutoff values from a dilution curve with non-linear fit in GraphPad Prism v.9.5.1. Normalization for batch effect was addressed by the inclusion of a positive control pool generated from pre-2022 controls with prior smallpox vaccination. Sera with titers below the limit of detection (50) were scored as 25. ### Statistical Methods Paired measurements in figures are analyzed by Wilcoxon matched-pairs signed rank test. Comparisons of multiple sample groups in figures are conducted by Kruskal-Wallis test with Dunn’s method for multiple comparisons. Linear mixed-effects regressions (LMER) were used to model neutralizing titer durability with repeated measurements. The *lme4* package (v.1.1-35.1) in R v.4.3.2 was used for all LMER calculations. Models were fit using the natural log transformation of the neutralizing titers. Both random intercepts only and random intercepts and slopes models were tested, but random intercepts only models were used as random intercepts and slopes models either failed to converge or had insufficient data for model fitting. Model predictions were generated using the *ggeffects* package (v.1.3.2). Half-lives were calculated by rearrangement of the LMER to its exponential decay form and used the predicted slope for days post dose 2. LMER model parameters for each instance are detailed in the Supplementary Materials. Correlation testing, paired measurements, and multiple comparison testing were all conducted in GraphPad Prism v.10.0.3. For all figures, p-value <0.05 is *, <0.01 is **, and <0.001 is \***|. ## Results ### The New York City Observational Study of Mpox Immunity We report here on 159 MVA-BN vaccinees with or without a prior history of smallpox vaccination (Table 1) and nine mpox convalescent individuals (Supplementary Table S1). The remaining study participants are excluded due to missing data or visits. Study visits are described in Figure 1; participants could enroll at any time point. Time points include a baseline (V1), a visit between vaccine doses (V2), and three visits following receipt of the second vaccine dose: median visit dates of ∼3 weeks post dose two (V3), ∼3 months post dose two (V4), and ∼9-10 months post dose two (V5). The majority of participants were younger than 50 years old and identified as male and a member of the lesbian, gay, bisexual, transgender, and queer+ community (LGBTQ+), in line with the groups recommended to receive vaccination as outlined by the CDC19. Of note, the median age for vaccinees with a history of smallpox vaccination was 62, as compared to 33 for those without prior smallpox vaccination. Nearly a third of the participants had received a prior smallpox vaccination (referred to herein as experienced participants). Approximately half of those experienced participants were PWH, while only ∼15% of naïve participants were PWH. Due to the EUA for ID administration, OSMI includes a range of dosing combinations: SC-SC, SC-ID, ID-SC, and ID-ID. SC-ID was the most common in the study (∼33% of experienced participants and ∼46% of naïve participants). View this table: [Table 1:](http://medrxiv.org/content/early/2024/01/29/2024.01.28.24301893/T1) Table 1: Demographics of the NYC Observational Study of Mpox Immunity ### Peak MPXV neutralization titers are comparable for naïve and experienced MVA-BN vaccinees but differ significantly in durability To measure the neutralizing titers of OSMI participants, a fluorescence-based microneutralization assay was developed using authentic clade IIb MPXV. Pre-outbreak samples from VACV-experienced individuals have significantly higher neutralizing titers at baseline (Fig. 2A, pre-2022 controls), in line with prior work showing long-lasting durability for orthopoxvirus immunity8–12. Aggregate data show no statistically significant difference between naïve and experienced participants at V3 (geometric mean titers [GMTs] of 95 versus 127, respectively), highlighting similar peak neutralizing titers (Fig. 2A). However, by V4, there is a significant difference between the two groups (GMTs of 45 for naïve versus 111 for experienced). This significant difference continues at V5 with 81% of VACV-naïve vaccinees below the positivity threshold, compared to only 14% of VACV-experienced vaccinees (GMTs of 24 versus 85, respectively). Nine mpox-convalescent individuals are included as another control group, some of whom had received at least one dose of MVA-BN (Supplementary Table S1). At ∼1 year after symptom onset, these participants have GMTs greater than or equal to peak titers for vaccinees, suggesting a more durable neutralizing antibody response post MPXV infection as compared to MVA-BN vaccination. These temporal patterns of neutralizing antibodies over one year are also observed for anti-MPXV H3 IgG binding antibody titers by ELISA, although a lower proportion of VACV-naïve MVA-BN vaccinees (50%) are the below the limit of detection for one-year H3-binding antibody (Supplementary Fig. S1A). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/01/29/2024.01.28.24301893/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2024/01/29/2024.01.28.24301893/F2) Figure 2: The MPXV neutralizing titers of naïve individuals wane at a significantly faster rate than those of experienced individuals A) Aggregate mpox virus (MPXV) neutralizing titer data for each study visit sorted by prior smallpox vaccination status. Colored/black bars on each group indicate the geometric mean titer. Serum neutralizing titers were measured by a fluorescence-based microneutralization assay using authentic clade IIb MPXV. Pre-2022 controls come from the New York University Langone Vaccine Center Biorepository. Convalescent (conv) participants are sorted by whether they received MVA-BN. B) Longitudinal neutralizing titers from naïve participants across all five study visits. Bolded line indicates the mean neutralizing titer for participants at each time point. C) Longitudinal neutralizing titers from experienced participants across all five study visits. Bolded line indicates the mean neutralizing titer for participants at each time point. Horizontal, black dashed line in all panels indicates the positivity threshold for neutralization (ID50 = 33.7) as based on the pre-2022 negative controls (mean of controls plus two times the standard deviation). Statistical testing conducted by Kruskal-Wallis test with Dunn’s method for multiple comparisons for panel A and Wilcoxon matched-pairs signed rank test for panels B and C. Exp, experienced vaccinees (prior history of smallpox vaccination); % neg, percent of samples below the positivity threshold. ### HIV status has no significant effect on responses to MVA-BN in people with CD4 counts over 250 cells/mm3 One of the original study objectives was to determine in PWH whether the EUA dose-sparing ID route was immunogenically comparable to the licensed full-dose SC route, as prior studies of MVA-BN vaccination in PWH only examined the approved SC route20,21. All of our participants with HIV had CD4+ T cell counts over 250 and most were over 500 (Supplementary Table S2). Combining all routes of vaccination, neutralizing titers are similar between HIV-uninfected participants and PWH in both the naïve and experienced groups (Supplementary Fig. S2). When focusing only on those that received at least one ID dose, a similar result is found (Supplementary Fig. S3). We additionally examined the effect of HIV status in a linear mixed-effects regression and similarly found no significant effect on titers in VACV-naïve (Supplementary Table S3) or VACV-experienced participants (Supplementary Table S4). Lastly, using a linear mixed-effects model, we tested whether CD4 counts had a significant effect on titers and found no such effect in naïve (Supplementary Table S5) or experienced participants (Supplementary Table S6). ### Neutralizing titers decay significantly faster in naïve vaccinees compared to experienced vaccinees To further leverage the longitudinal nature of the OSMI study, we separately examined participants with samples from consecutive visits (Fig. 2B and C). Following the first dose of MVA-BN, naïve participants already had significantly higher neutralizing titers (Fig. 2B). This significant rise continued after the second dose, with an observed peak at V3. Subsequent visits each showed significant declines. For experienced individuals (Fig. 2C), there were insufficient paired samples to gauge the effects of the first dose, but following dose two, titers did not begin to decline until after V4. A similar pattern was seen with anti-MPXV H3 IgG titers (Supplementary Fig. S1B and C). To statistically evaluate the rate of change in each group, paired fold changes in MPXV neutralizing titers for V3 versus V4 and V4 versus V5 were compared for naïve versus experienced individuals. In both instances, naïve participants have a significantly greater loss of titers from one visit to the next (Supplementary Fig. S4). This was further borne out by modeling titers over time using a linear mixed-effects regression (Supplementary Tables S3 and S4). For naïve participants, neutralizing titers were significantly dependent on dosing interval and days post dose two (model parameters can be found in Supplementary Table S7), while neutralizing titers in experienced participants only significantly depended upon days post dose two (Supplementary Table S8). Using these models, the half-life of neutralizing antibodies is predicted to be 168 days (95% C.I.: 147 to 197 days) in naïve individuals, regardless of dosing interval. Participants that received the licensed 1-month dosing interval lost titer positivity by 160 days post dose two (Fig. 3A). The time to negativity increased by ∼1 month for each additional month between MVA-BN doses (Fig. 3A inset table). This is likely driven, in part, by the significant positive correlation of peak MPXV neutralizing titers with dosing interval (Supplementary Fig. 5). Antibody decline is far slower in VACV-experienced MVA-BN vaccinees (Fig. 3B) with a predicted half-life of 387 days (95% C.I.: 289 to 600 days). The predicted time to cross the neutralizing titer positivity threshold is >350 days, i.e., beyond the available data. Together these data highlight the difference in the longevity of serum neutralization activity for the two-dose MVA-BN regimen in VACV-naïve people versus VACV-experienced people. ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/01/29/2024.01.28.24301893/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2024/01/29/2024.01.28.24301893/F3) Figure 3: Modeling of neutralizing titer durability shows naïve individuals revert to negative titers in under a year, while experienced individuals remain positive beyond a year A) Linear mixed-effects regression of naïve participants using all data points after the second dose of MVA-BN. Regressions of natural log-transformed neutralizing titers use days post dose 2 and dosing interval as fixed effects and account for random subject effects by allowing for random intercepts. Colored regression lines with transparent ribbons indicate the predicted titers with 95% confidence interval. Model parameters are listed in Supplementary Table S7. B) Linear mixed-effects regression of experienced participants using all data points after the second dose of MVA-BN. Regression of natural log-transformed neutralizing titers uses days post dose 2 as a fixed effect and accounts for random subject effects by allowing for random intercepts. The solid black regression line with transparent ribbon indicates the predicted titers with 95% confidence interval. Model parameters are listed in Supplementary Table S8. Horizontal, black dashed line in each panel indicates the positivity threshold for neutralization (Fig. 2). ### IgG responses to MPXV A35 and H3 are immunodominant regardless of smallpox vaccination history, but have lower avidity in naïve vaccinees To further characterize the serological responses of the naïve and experienced groups, a subset of participants with multiple study visits from each group was created. The sera subsets have neutralizing titers similar to the larger study groups (Supplementary Fig. S6). Using a multiplexed immunoassay, IgG binding titers and avidity for the following eight MPXV antigens (Table 2) were measured: A29, A30, A35, B16, B21, E8, H3, and L1. These surface antigens were selected as known protective targets22–29 or potential neutralizing targets based on previous reports30–32. IgG avidity was measured in this assay using an additional incubation step with 2M ammonium thiocyanate following serum incubation. We found that the H3 and A35 viral proteins were immunodominant in both naïve and experienced individuals across all time points post-dose two (Supplementary Fig. S7A). Binding titers for H3 and A35 were also significantly correlated with MPXV neutralization in both groups across multiple time points (Supplementary Fig. S7B). As the VACV homologs of H3 and A35 have previously been shown to be protective antibody targets in orthopoxvirus infections22–24, we decided to focus on these two antigens for our analysis. View this table: [Table 2:](http://medrxiv.org/content/early/2024/01/29/2024.01.28.24301893/T2) Table 2: Eight MPXV proteins used in the multiplexed immunoassay for antibody binding and avidity: their location in virions and function in viral infection Similar to MPXV neutralizing titers, both anti-H3 (Fig. 4A) and anti-A35 (Fig. 4B) IgG reach comparable peak titers in naïve and experienced individuals. IgG titers against each antigen also decline faster in naïve participants as compared to experienced vaccinees. Anti-H3 IgG begins to wane after V3 in both groups (Fig. 4C and 4D), while anti-A35 IgG begin to wane after V3 in naïve individuals (Fig. 4E), but only after V4 in experienced individuals (Fig. 4F). The timing of A35 waning more closely matches the waning of neutralizing titers in experienced vaccinees. We also measured avidity for anti-H3 and -A35 IgG. Aggregate data show significantly higher IgG avidity against H3 (Fig. 5A) and A35 (Fig. 5B) in the VACV-experienced group across all time points post dose two. Surprisingly, longitudinal samples had relatively limited changes in anti-H3 IgG avidity for VACV-naïve vaccinees (Fig. 5C). However, anti-H3 IgG avidity significantly declines from V3 to V4 and V4 to V5 in the experienced group (Fig. 5D), indicating the generation of *de novo* low avidity responses that reduce the bulk avidity in the sera. Changes in the avidity of anti-A35 IgG are more muted but of a similar direction (Fig. 5E and 5F). Taken together these data suggest similar B cell specificities (immunodominance) for naïve and experienced MVA-BN vaccinees, but a dramatic difference with avidity being much higher in VACV-experienced MVA-BN vaccinees. We observed no clear evidence of ongoing affinity maturation in samples post dose two in either naïve or experienced individuals. ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/01/29/2024.01.28.24301893/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2024/01/29/2024.01.28.24301893/F4) Figure 4: Anti-MPXV H3 and -A35 IgG reach comparable peak titers in naïve and experienced participants, but wane more quickly in naïve participants A) Aggregate anti-MPXV H3 IgG titers as measured by multiplexed immunoassay. Titers are reported as AUC from two serum dilutions. B) Aggregate anti-MPXV A35 IgG titers as measured by multiplexed immunoassay. Titers are reported as AUC from two serum dilutions. C) Longitudinal anti-MPXV H3 IgG titers for naïve participants at V1 and V3-5 as measured by multiplexed immunoassay. Titers are expressed as AUC from two serum dilutions. D) Same as C, but for experienced participants across V3-5. E) Longitudinal anti-MPXV A35 IgG titers for naïve participants at V1 and V3-5 as measured by multiplexed immunoassay. Titers are expressed as AUC from two serum dilutions. F) Same as E, but for experienced participants across V3-5. In panels A and B, colored/black bars indicate the geometric mean. For panels C-F, bolded line indicates the mean IgG titer for participants at each time point. Black, dashed line in all panels is positivity threshold for binding as determined by pre-2022 negative controls (same samples and method as in Fig 2). Statistical testing conducted by Kruskal-Wallis test with Dunn’s method for multiple comparisons for panels A and B, and by Wilcoxon matched-pairs signed rank test for panels C-F. ![Figure 5:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/01/29/2024.01.28.24301893/F5.medium.gif) [Figure 5:](http://medrxiv.org/content/early/2024/01/29/2024.01.28.24301893/F5) Figure 5: Experienced vaccinees generate higher avidity IgG against MPXV H3 and A35 A) Aggregate avidity indices for anti-MPXV H3 IgG. B) Aggregate avidity indices for anti-MPXV A35 IgG. C) Longitudinal avidity indices for anti-MPXV H3 IgG antibodies for V3-5 of naïve participants. D) Same as C, but for experienced participants. E) Longitudinal avidity indices for anti-MPXV A35 IgG antibodies for V3-5 of naïve participants. F) Same as E, but for experienced participants. Avidity was measured in a multiplexed immunoassay using a 2M ammonium thiocyanate (NH4SCN) wash and expressed as the ratio of the AUC from the NH4SCN condition over the AUC from a PBS control condition. In panels A and B, colored/black bars indicate the mean. Statistical testing conducted by Kruskal-Wallis test with Dunn’s method for multiple comparisons for panels A and B, and by Wilcoxon matched-pairs signed rank test for panels C-F. Exp, experienced vaccinees (prior history of smallpox vaccination). ## Discussion We report here an analysis through one year of the antibody specificity and avidity against MPXV following the two-dose MVA-BN vaccination regimen. Our study is ongoing but the current data show that serological responses to MVA-BN are limited in their durability. Prior work has highlighted the limited duration of neutralizing titers against VACV or MPXV in sera from MVA-BN vaccinated people13,14, and that is confirmed in our own cohort. We identified immunodominant MPXV proteins and found that the IgG response to those proteins has low avidity in VACV-naïve MVA-BN recipients in comparison to VACV-experienced MVA-BN recipients and compared to what might be expected33–35. Our data offer reassurances that the immunity generated by ID administration of one-fifth dose MVA-BN is comparable between PWH and HIV-uninfected individuals. This information is particularly timely in light of the ongoing mpox clade I outbreak in the Democratic Republic of the Congo, where vaccination roll-out has been extremely limited and dose-sparing approaches could be appropriate36,37. MVA-BN would be expected to induce comparable immunity against MPXV clades I and II based on their high sequence homology, but this assumption needs confirmation. The differences we see between VACV-naïve and VACV-experienced MVA-BN vaccinees may arise from a number of sources. While the VACV-experienced group received a dose of replicating vaccine (vaccinia), the naïve group received only the non-replicating MVA-BN. These differences in viral replication are likely to generate different inflammatory states, which we have recently shown durably impacts the CD4+ T cell memory response38. The two groups also differ in quantity of vaccine exposures (three versus two doses) as well as the timing of first vaccine exposure (childhood versus adulthood). To the latter point, however, prior work has shown that first-time vaccination with 1st or 2nd generation VACV vaccines in adulthood yields more durable neutralizing titers than were observed here in VACV-naïve adults receiving MVA-BN (∼10 years versus <1 year, respectively)12. This would argue that VACV vaccine replication is a significant contributor to the durability of immunity. Lastly, as VACV-specific memory B cells and memory T cells are known to persist for decades8,9,11, anamnestic responses are likely driving much of the response to MVA-BN in VACV-experienced MVA-BN recipients, resulting in the observed higher durability and higher avidity of serum IgG relative to the naïve MVA-BN vaccinees. The average mpox incubation period of just over a week39 may be short enough that memory B cells and memory T cells can activate and protect an exposed human even in the absence of circulating neutralizing antibodies. How waning of antibodies impacts transmission is also an open question as recent work has revealed that viral DNA and culturable virus is present in infected individuals as early as 4 days prior to symptom onset40. Our finding here that a longer dosing interval increases neutralizing titer durability among naïve vaccinees differs from our previous work where we reported that there was no significant effect of interval on durability of anti-MPXV H3 IgG titers in naïve individuals7. We do not consider these findings to be at odds as we and others have shown that there is an imperfect correlation between anti-H3 titers and orthopoxvirus neutralization41,42. It is interesting to note that there is no significant effect of interval on neutralizing titers in VACV-experienced individuals. A possible explanation is that the second dose of MVA-BN is unnecessary in this group. Previous work points to this43. The delayed rollout of second doses during the 2022 outbreak offered a unique opportunity to begin studying varied dosing schedules, but controlled studies would be necessary to identify the ideal dose interval. As an observational study, OSMI is limited in a number of ways. The observational nature of the study as well as the accelerated time span of the 2022 mpox outbreak created difficulties in obtaining baseline samples for many of the participants in OSMI, especially the smaller group of individuals with prior VACV vaccination. Likewise, the number of participants that received a SC first dose and an ID second dose far outweigh the other vaccination route combinations due to the nature of the vaccination campaigns and dose availability. We also do not report on cellular immunity here, but work is ongoing in our laboratories to use the sample set afforded by OSMI to better understand the role of cellular immunity in MVA-BN responses. NYC OSMI is an observational study initiated in a public health emergency with specimen collection timepoint windows that are wider than in a typical vaccine clinical trial. Categorization of subjects as VACV-naïve versus -experienced is not a perfect undertaking given the lack of record availability for most participants. The categorization of subjects into route of immunization groups (SC-SC, ID-ID, ID-SC, and SC-ID) was based on immunization records whenever possible, but sometimes relied on participant descriptions and knowledge. An additional limitation of our work is that the eight MPXV proteins chosen for antibody binding analyses are a small subset of known neutralization targets for orthopoxviruses. Directions for future work include further study of the magnitudes, specificities, qualities, and durability of memory B cells and memory T cells following the two-dose MVA-BN regimen. Further study of the benefit, or lack thereof, of the second MVA-BN dose in VACV-experienced people is needed. Additional study of the ability of clade I MPXV to be neutralized by sera from MVA-BN-vaccinated people is needed. The non-neutralizing antiviral functions of antibodies should be studied; e.g., ADCC. It seems prudent to study the immune responses of third dose boosters in VACV-naïve people who are a year or more after their MVA-BN primary series. It will be important for real-world efficacy studies to assess the incidence and timing of post-vaccination breakthrough infections. A critical unanswered question is whether the memory cellular responses induced by the MVA-BN two-dose regimen in VACV-naïve vaccinees is sufficient to protect against mpox disease at timepoints when circulating neutralizing antibody is no longer detectable. Overall, we have shown that the two-dose MVA-BN regimen in naïve individuals produces low avidity, nondurable serological responses that significantly differ from the response in individuals with prior smallpox vaccination. While MVA-BN offers a crucial vaccine option that would otherwise not be available to most immunocompromised individuals, it is important to better understand its current limitations. An ideal future mpox vaccine would combine the safety advantages of the non-replicating MVA-BN with the robust, durable protective immunity of replicating VACV vaccines. It is currently unclear how MVA-BN vaccination can be augmented to bridge this divide. Future studies that we and others are undertaking of MVA-BN cellular immunity may offer clues as to how to improve immune responses, whether that is through a molecular adjuvant, additional doses, higher doses, altered dosing intervals, or even virologic changes to MVA-BN itself. This is particularly important as mpox outbreaks look poised to continue into the future. The findings here regarding antibody specificity and quality may offer insights for efforts to produce next-generation mpox-specific vaccines44–46. ## Supporting information Supplemental Materials [[supplements/301893_file02.pdf]](pending:yes) ## Data Availability All data produced in the present study are available upon reasonable request to the authors. ## Conflicts of Interest M.J.M. reported potential competing interests: laboratory research and clinical trials contracts for vaccines or MAB with Lilly, Pfizer, and Sanofi; research grant funding from USG/HHS/NIH for vaccine and MAB clinical trials; personal fees for Scientific Advisory Board service from Hillevax, Merck, Meissa Vaccines, Inc. and Pfizer. ## Acknowledgements Most importantly, the authors thank the NYC OSMI study participants for their altruism and volunteerism, without whom we would have no study. The authors wish to acknowledge the full NYC OSMI Study Group, which includes, in addition to the byline authors, the following colleagues: Abdul Abdulai; Jaqueline Callahan, RN; Ellie Carmody, MD, MPH; Tamia Davis, NP; Amanda Dontino; Aimee Edwin, RN; Celia Engelson, NP; Olivia Frank, MPH; Emily Geesey; Shelby Goins; Victoria Guerra, RN; Sarah Haiken; Trishala Karmacharya; Dorothy Knutsen, MD; Irma Noriega, NP; Samuel Nweke; Lalitha Parameswaran, MD, MPH; Gurchetan Randhawa, MD; Miguel Rodriguez; Pamela Suman; Meron Tasissa; Kathryn Swindell, NP; Julia Wagner, MPH; Jimmy P. Wilson; Samantha Yip, RN; Lisa Zhao. The authors would like to thank Dr. Meike Dittmann and Austin Schinlever for their assistance in cultivating MPXV viral stocks. We also thank the NYU Office of Scientific Research Animal Biosafety Level 3 staff, Drs. Ludovic Desvignes, Dominick Papandrea, and Alison Gilchrist, for their support of this work. Financial support was provided by the Blavatnik Family Foundation, the National Institutes of Health (NIH) grants no. AI148574 and 75N93021C00014 (to M.J.M.), the NYC Department of Health and Mental Hygiene, and NYU Grossman School of Medicine institutional support. * Received January 28, 2024. * Revision received January 28, 2024. * Accepted January 29, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution 4.0 International), CC BY 4.0, as described at [http://creativecommons.org/licenses/by/4.0/](http://creativecommons.org/licenses/by/4.0/) ## References 1. 1.2022-23 Mpox (Monkeypox) Outbreak: Global Trends. [https://worldhealthorg.shinyapps.io/mpx_global/](https://worldhealthorg.shinyapps.io/mpx_global/). 2. 2.2022 Outbreak Cases and Data | Mpox | Poxvirus | CDC. [https://www.cdc.gov/poxvirus/mpox/response/2022/index.html](https://www.cdc.gov/poxvirus/mpox/response/2022/index.html). 3. 3.Allan-Blitz, L. T. et al. A Position Statement on Mpox as a Sexually Transmitted Disease. Clinical Infectious Diseases 76, 1508–1512 (2023). 4. 4.Bunge, E. M. et al. The changing epidemiology of human monkeypox—A potential threat? A systematic review. PLoS Negl Trop Dis 16, e0010141 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pntd.0010141&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35148313&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 5. 5.Interim Clinical Considerations for Use of JYNNEOS and ACAM2000 Vaccines during the 2022 U.S. Mpox Outbreak | Mpox | Poxvirus | CDC. [https://www.cdc.gov/poxvirus/mpox/clinicians/vaccines/vaccine-considerations.html](https://www.cdc.gov/poxvirus/mpox/clinicians/vaccines/vaccine-considerations.html). 6. 6.Frey, S. E. et al. Comparison of lyophilized versus liquid modified vaccinia Ankara (MVA) formulations and subcutaneous versus intradermal routes of administration in healthy vaccinia-naïve subjects. Vaccine 33, 5225–5234 (2015). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2015.06.075&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26143613&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 7. 7.Kottkamp, A. C. et al. Antibody Titers against Mpox Virus after Vaccination. New England Journal of Medicine 389, 2299–2301 (2023). 8. 8.Hammarlund, E. et al. Duration of antiviral immunity after smallpox vaccination. Nature Medicine 2003 9:9 9, 1131–1137 (2003). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm917&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12925846&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000185061600024&link_type=ISI) 9. 9.Amanna, I. J., Carlson, N. E. & Slifka, M. K. Duration of Humoral Immunity to Common Viral and Vaccine Antigens. New England Journal of Medicine 357, 1903–1915 (2007). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa066092&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17989383&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000250732500005&link_type=ISI) 10. 10.Taub, D. D. et al. Immunity from Smallpox Vaccine Persists for Decades: A Longitudinal Study. Am J Med 121, 1058–1064 (2008). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.amjmed.2008.08.019&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19028201&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000261985100025&link_type=ISI) 11. 11.Crotty, S. et al. Cutting Edge: Long-Term B Cell Memory in Humans after Smallpox Vaccination. The Journal of Immunology 171, 4969–4973 (2003). 12. 12.Slike, B. M., Creegan, M., Marovich, M. & Ngauy, V. Humoral Immunity to Primary Smallpox Vaccination: Impact of Childhood versus Adult Immunization on Vaccinia Vector Vaccine Development in Military Populations. PLoS One 12, e0169247 (2017). 13. 13.Ilchmann, H. et al. One- and Two-Dose Vaccinations With Modified Vaccinia Ankara-Bavarian Nordic Induce Durable B-Cell Memory Responses Comparable to Replicating Smallpox Vaccines. J Infect Dis 227, 1203–1213 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiac455&link_type=DOI) 14. 14.Priyamvada, L. et al. Serological responses to the MVA-based JYNNEOS monkeypox vaccine in a cohort of participants from the Democratic Republic of Congo. Vaccine 40, 7321–7327 (2022). 15. 15.Cohn, H. et al. Mpox vaccine and infection-driven human immune signatures: an immunological analysis of an observational study. Lancet Infect Dis , (2023). 16. 16.Otter, A. D. et al. Monkeypox virus-infected individuals mount comparable humoral immune responses as Smallpox-vaccinated individuals. Nature Communications 2023 14:1 14, 1–14 (2023). 17. 17.Zaeck, L. M. et al. Low levels of monkeypox virus-neutralizing antibodies after MVA-BN vaccination in healthy individuals. Nature Medicine 2022 1–9 (2022) doi:10.1038/s41591-022-02090-w. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-022-02090-w&link_type=DOI) 18. 18.Rao, A. K. et al. Use of JYNNEOS (Smallpox and Monkeypox Vaccine, Live, Nonreplicating) for Preexposure Vaccination of Persons at Risk for Occupational Exposure to Orthopoxviruses: Recommendations of the Advisory Committee on Immunization Practices — United States, 2022. MMWR Morb Mortal Wkly Rep 71, 734–742 (2022). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.15585/mmwr.mm7122e1&link_type=DOI) 19. 19.Mpox Vaccine Recommendations | Mpox | Poxvirus | CDC. [https://www.cdc.gov/poxvirus/mpox/vaccines/vaccine-recommendations.html](https://www.cdc.gov/poxvirus/mpox/vaccines/vaccine-recommendations.html). 20. 20.Greenberg, R. N. et al. Safety, Immunogenicity, and Surrogate Markers of Clinical Efficacy for Modified Vaccinia Ankara as a Smallpox Vaccine in HIV-Infected Subjects. J Infect Dis 207, 749–758 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jis753&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23225902&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 21. 21.Overton, E. T. et al. A randomized phase II trial to compare safety and immunogenicity of the MVA-BN smallpox vaccine at various doses in adults with a history of AIDS. Vaccine 38, 2600–2607 (2020). 22. 22.McCausland, M. M. et al. Combination therapy of vaccinia virus infection with human anti-H3 and anti-B5 monoclonal antibodies in a small animal model. Antivir Ther 15, 661–675 (2010). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3851/IMP1573&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20587859&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 23. 23.Davies, D. H. et al. Vaccinia Virus H3L Envelope Protein Is a Major Target of Neutralizing Antibodies in Humans and Elicits Protection against Lethal Challenge in Mice. J Virol 79, 11724–11733 (2005). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjExOiI3OS8xOC8xMTcyNCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAxLzI5LzIwMjQuMDEuMjguMjQzMDE4OTMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 24. 24.Chen, Z. et al. Characterization of Chimpanzee/Human Monoclonal Antibodies to Vaccinia Virus A33 Glycoprotein and Its Variola Virus Homolog In Vitro and in a Vaccinia Virus Mouse Protection Model. J Virol 81, 8989–8995 (2007). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjEwOiI4MS8xNy84OTg5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDEvMjkvMjAyNC4wMS4yOC4yNDMwMTg5My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 25. 25.Diesterbeck, U. S. et al. Characterization of an in vivo neutralizing anti-vaccinia virus d8 single-chain fragment variable (Scfv) from a human anti-vaccinia virus-specific recombinant library. Vaccines (Basel) 9, 1308 (2021). 26. 26.Kaever, T. et al. Linear Epitopes in Vaccinia Virus A27 Are Targets of Protective Antibodies Induced by Vaccination against Smallpox. J Virol 90, 4334–4345 (2016). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjk6IjkwLzkvNDMzNCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAxLzI5LzIwMjQuMDEuMjguMjQzMDE4OTMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 27. 27.Nelson, G. E., Sisler, J. R., Chandran, D. & Moss, B. Vaccinia virus entry/fusion complex subunit A28 is a target of neutralizing and protective antibodies. Virology 380, 394–401 (2008). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.virol.2008.08.009&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18789472&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 28. 28.Gilchuk, I. et al. Cross-Neutralizing and Protective Human Antibody Specificities to Poxvirus Infections. Cell 167, 684–694.e9 (2016). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2016.09.049&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27768891&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 29. 29.Galmiche, M. C., Goenaga, J., Wittek, R. & Rindisbacher, L. Neutralizing and Protective Antibodies Directed against Vaccinia Virus Envelope Antigens. Virology 254, 71–80 (1999). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1006/viro.1998.9516&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9927575&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000078622700008&link_type=ISI) 30. 30.Duke-Cohan, J. S. et al. The heterogeneity of human antibody responses to vaccinia virus revealed through use of focused protein arrays. Vaccine 27, 1154–1165 (2009). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.vaccine.2008.12.035&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19146908&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 31. 31.Chiu, W.-L. & Chang, W. Vaccinia Virus J1R Protein: a Viral Membrane Protein That Is Essential for Virion Morphogenesis. J Virol 76, 9575–9587 (2002). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjEwOiI3Ni8xOS85NTc1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDEvMjkvMjAyNC4wMS4yOC4yNDMwMTg5My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 32. 32.Hammarlund, E. et al. Multiple diagnostic techniques identify previously vaccinated individuals with protective immunity against monkeypox. Nature Medicine 2005 11:9 11, 1005–1011 (2005). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm1273&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16086024&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000231724700030&link_type=ISI) 33. 33.Puschnik, A. et al. Correlation between Dengue-Specific Neutralizing Antibodies and Serum Avidity in Primary and Secondary Dengue Virus 3 Natural Infections in Humans. PLoS Negl Trop Dis 7, e2274 (2013). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pntd.0002274&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23785536&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom) 34. 34.Nakagama, Y. et al. Antibody Avidity Maturation Following Recovery From Infection or the Booster Vaccination Grants Breadth of SARS-CoV-2 Neutralizing Capacity. J Infect Dis 227, 780–787 (2023). 35. 35.Wec, A. Z. et al. Longitudinal dynamics of the human B cell response to the yellow fever 17D vaccine. Proc Natl Acad Sci U S A 117, 6675–6685 (2020). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTE3LzEyLzY2NzUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMS8yOS8yMDI0LjAxLjI4LjI0MzAxODkzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 36. 36.Health Alert Network (HAN) - 00501 | Mpox Caused by Human-to-Human Transmission of Monkeypox Virus with Geographic Spread in the Democratic Republic of the Congo. [https://emergency.cdc.gov/han/2023/han00501.asp](https://emergency.cdc.gov/han/2023/han00501.asp). 37. 37.Mpox (monkeypox)-Democratic Republic of the Congo. [https://www.who.int/emergencies/disease-outbreak-news/item/2023-DON493](https://www.who.int/emergencies/disease-outbreak-news/item/2023-DON493). 38. 38.Gray-Gaillard, S. L. et al. Inflammation durably imprints memory CD4+ T cells. bioRxiv 2022.11.15.516351 (2023) doi:10.1101/2022.11.15.516351. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMi4xMS4xNS41MTYzNTF2MiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAxLzI5LzIwMjQuMDEuMjguMjQzMDE4OTMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 39. 39.Madewell, Z. J. et al. Serial Interval and Incubation Period Estimates of Monkeypox Virus Infection in 12 Jurisdictions, United States, May–August 2022 - Volume 29, Number 4— April 2023 - Emerging Infectious Diseases journal - CDC. Emerg Infect Dis 29, 818–821 (2023). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3201/eid2904.221622&link_type=DOI) 40. 40.Brosius, I. et al. Presymptomatic viral shedding in high-risk mpox contacts: A prospective cohort study. J Med Virol 95, e28769 (2023). 41. 41.Benhnia, M. R.-E.-I. et al. Redundancy and Plasticity of Neutralizing Antibody Responses Are Cornerstone Attributes of the Human Immune Response to the Smallpox Vaccine. J Virol 82, 3751–3768 (2008). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjk6IjgyLzcvMzc1MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAxLzI5LzIwMjQuMDEuMjguMjQzMDE4OTMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 42. 42.Frey, S. E. et al. Human Antibody Responses Following Vaccinia Immunization Using Protein Microarrays and Correlation With Cell-Mediated Immunity and Antibody-Dependent Cellular Cytotoxicity Responses. J Infect Dis 224, 1372–1382 (2021). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiab111&link_type=DOI) 43. 43.Sammartino, J. C. et al. Characterization of immune response against monkeypox virus in cohorts of infected patients, historic and newly vaccinated subjects. J Med Virol 95, e28778 (2023). 44. 44.Freyn, A. W. et al. An mpox virus mRNA-lipid nanoparticle vaccine confers protection against lethal orthopoxviral challenge. Sci Transl Med 15, eadg3540 (2023). 45. 45.Hou, F. et al. mRNA vaccines encoding fusion proteins of monkeypox virus antigens protect mice from vaccinia virus challenge. Nature Communications 2023 14:1 14, 1–10 (2023). 46. 46.Zeng, J. et al. Mpox multi-antigen mRNA vaccine candidates by a simplified manufacturing strategy afford efficient protection against lethal orthopoxvirus challenge. Emerg Microbes Infect 12, (2023). 47. 47.Colamonici, O. R., Domanski, P., Sweitzer, S. M., Larner, A. & Buller, R. M. L. Vaccinia Virus B18R Gene Encodes a Type I Interferon-binding Protein That Blocks Interferon α Transmembrane Signaling. Journal of Biological Chemistry 270, 15974–15978 (1995). [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamJjIjtzOjU6InJlc2lkIjtzOjEyOiIyNzAvMjcvMTU5NzQiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMS8yOS8yMDI0LjAxLjI4LjI0MzAxODkzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 48. 48.Szajner, P., Jaffe, H., Weisberg, A. S. & Moss, B. A complex of seven vaccinia virus proteins conserved in all chordopoxviruses is required for the association of membranes and viroplasm to form immature virions. Virology 330, 447–459 (2004). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.virol.2004.10.008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15567438&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F01%2F29%2F2024.01.28.24301893.atom)