ABSTRACT
SARS-CoV-2 antibody levels associated with reduced hospitalization risk remain undefined. Our outpatient COVID-19 convalescent plasma (CCP), placebo-controlled trial observed SARS-CoV-2 antibody levels decreasing 22-fold from matched donor units into post-transfusion seronegative recipients. Unvaccinated recipients were jointly stratified by a) early or late transfusion (≤ 5 or >5 days from symptom onset) and b) high or low post-transfusion SARS-CoV-2 antibody levels (< or ≥ geometric mean). Early treatment with high post-transfusion antibody levels reduced hospitalization risk-0/102 (0%) compared to all other CCP recipients-17/370 (4.6%; Fisher exact p=0.03) and to all control plasma recipients-35/461 (7.6%; Fisher exact p=0.001). A similar donor upper/lower antibody level and early late transfusion stratified analyses indicated significant hospital risk reduction. Pre-transfusion nasal viral loads were similar in CCP and control recipients regardless of hospitalization outcome. Therapeutic CCP should comprise the upper 30% of donor antibody levels to provide effective outpatient use for immunocompromised and immunocompetent outpatients.
INTRODUCTION
The threshold of antibody correlating with reduced severe disease progression varies and needs to be independently determined for diverse infectious diseases, from protozoans, like Plasmodium falciparum1,2 to viruses, like SARS-CoV-23,4. Correlates of protection for antibody levels demarcating vaccine efficacy for COVID-19 hospitalization risk reduction have been represented in relation to COVID-19 convalescent plasma (CCP) equivalents5-7. Likewise, a model8 utilizing vaccine, monoclonal antibody, and CCP data from clinical trials was created based on the reference CCP equivalents for 50% hospitalization risk reduction9-11. Monoclonal antibody outpatient randomized controlled trials (RCTs) demonstrate 50%-80% efficacy in hospital risk reduction12-15. However, the same monoclonal antibody doses failed to show disease progression reduction in COVID-19 inpatients14,16-18. Like the monoclonal antibodies, intravenous remdesivir demonstrated greater efficacy in outpatients versus inpatients19,20. In summary, both early timing and dose of antiviral antibodies and drugs matter for outpatient reduction of hospitalization risk.
Convalescent persons recovering from pre-Alpha period COVID-19 have varying antibody levels spanning a 3-log10 range from undetectable antibody to positive after a 3 to 5 thousand dilution21. Individuals with both boosted vaccines and recent COVID-19 (i.e., hybrid immunity) have considerably higher antibody levels (i.e., detectable even over 50,000 inverse dilutional geometric mean titers) than either individuals with antibodies from infection or vaccination alone 22,23. Our CCP outpatient treatment within 9 days of symptom onset, showed greater than 50% hospitalization relative risk reduction, which increased to more than 80% risk reduction (point estimate) when given within 5 days of symptom onset24. Hospitalized participants had the same donor unit range of SARS-CoV-2 antibody levels as those not hospitalized.
A prespecified analysis from the parent outpatient CCP RCT aimed to compare antibody levels in donor-recipient pairs to define dilutional decrease and to correlate both donor and recipient antibody levels to disease progression culminating in hospitalization. In this substudy, we analyzed the risk of hospitalization among unvaccinated COVID-19 outpatients, comparing control plasma recipients to four different CCP groups jointly stratified by early or late treatment (i.e., ≤ 5 or >5 days from symptom onset), and by high or low post-transfusion recipient or donor antibody levels (i.e., < or ≥ geometric mean, GM).
RESULTS
Trial population
At 23 sites throughout the USA, the previously reported outpatient CCP clinical trial (CSSC-004, NCT04373460) transfused 1181 COVID-19 outpatients over 16 months (June 3, 2020 to October 1, 2021), to show that receipt of CCP, compared to control plasma, reduced the risk of hospitalization for COVID-19 by 54%24. Written and signed informed consent was obtained from all participants. Hospitalizations occurred in 53 of 964 unvaccinated and 1 of 58 partially vaccinated participants. None of the 159 fully vaccinated individuals were hospitalized24. We focused our correlation of antibody level analysis and hospitalization on those unvaccinated with evaluable antibody data, for which 479 received control plasma and 472 CCP (Table 1). The unvaccinated participant’s mean age was 44, with more females than males, approximately 40% obese with BMI 30 or over, and approximately 40% with one or more pre-existing comorbidities for severe COVID-19 risk. Unvaccinated CCP recipients, both screen seropositive or seronegative were further jointly stratified by early (≤5 days of symptom onset) or late (>5 days of symptoms onset) time to transfusion as well as post-transfusion antibody levels (measured within 30 minutes after transfusion completion) in the upper or lower half of screened seronegative CCP recipients (Table 1).
Donor CCP and recipient screen and post-transfusion antibody levels in unvaccinated participants
Approximately 40% of all potential CCP study donors were excluded for low antibody levels, thus transfusion units represented the upper 60% of all convalescent plasma donors. Because of the greater correlation with virus neutralization antibody (nAb), we measured the plasma anti-Spike-Receptor Binding Domain (S-RBD) IgG antibodies levels by dilutional titer and the more precise area under the curve (AUC) on both the donors at collection and in over 5,000 recipient samples at pre-transfusion screen, post-transfusion and follow-up visits21. The anti-S-RBD IgG titer threshold for seronegative was 180 titer or below. The donor anti-S-RBD IgG GM titer of 6,678 and anti-S-RBD IgG AUC of 3,172 separated donor antibody levels’ upper and lower half (Fig. 1A). The donor control plasma was either collected in 2019 (211 units) or blood bank tested SARS-CoV-2 seronegative when collected in 2020-2021.
To experimentally determine the dilution factor associated with the single ∼200 mL CCP administration in 338 unvaccinated seronegative recipients, measurements of matched donor and recipient anti-S-RBD IgG AUC were performed. The matched transfusion donor antibody levels proportionately decreased 22-fold (approximately 5%) from the GM donor anti-S-RBD IgG 3286 AUC to recipient post-transfusion antibody levels at GM 147 AUC measured within 30 minutes of transfusion completion (Fig. 1B). The decrease was parallel such that matched donor antibody levels below 1000 AUC were also consistently below 150 AUC post-transfusion in recipients. Similarly, 15 seronegative hospitalized CCP recipients had post-transfusion antibody levels 19 times lower than their matched donors (Fig. 1C).
The unvaccinated study participants included a screen seropositive participant subset (199/951 (21%) (107 in the CCP and 92 in the control plasma groups), with 83% of the 199 seropositive participants already having pre-transfusion antibody levels above the GM 150 AUC for seronegative post-transfusion CCP recipients (Fig.1D). The pre-transfusion seropositive transfusion antibody levels were near 500 to 1000 GM anti-S-RBD IgG AUC which is 3 to 6 times lower than the donor anti-S-RBD IgG antibody levels of 3172 AUC. The low post-transfusion recipient antibodies (early or late) were predictably lower in frequency and GM AUC levels. Segregating the unvaccinated seropositive recipients by days from symptom onset to transfusion revealed a trend of a higher frequency of participants being seropositive after day 3, with higher GM antibody levels approximating 1000 anti-S-RBD IgG AUC, which was associated with the early development of virus-specific immunity also observed by Wolfel25 (Fig. 1E).
Recipient antibody level post-transfusion benchmarks of protection that prevents hospitalization
To correlate post-transfusion antibody levels among unvaccinated participants (both screen seronegative and seropositive) to hospital outcome, we stratified the post-transfusion anti-S-RBD IgG AUC by early or late treatment and high or low antibody levels for the CCP group (Fig. 2, Extended Table 1). The recipient antibody geometric mean of 150 AUC delineated high versus low antibody levels. Among the CCP participants treated early (symptom onset within 5 days) with high post-transfusion recipient antibody levels, there were no hospitalizations, 0/102 participants. In those unvaccinated CCP recipients treated early with low post-transfusion antibody levels (anti-S-RBD IgG AUC below 150 AUC), we observed 5/104 (5%) participant hospitalizations (Fig. 2). There were 12 total hospitalizations in the two groups receiving transfusions late—6/164 (3.7%) recipients with high antibody levels and 6/102 (6%) recipients with antibodies below anti-S-RBD IgG 150 AUC. We compared the proportion of hospitalizations of the early treatment with high post-transfusion antibody level CCP group with that of the other three CCP groups (17/270, 4.6%; Fishers exact p=0.03), the control plasma group (36/479, 7.5%; Fishers exact p=0.001), and the early control plasma group (24/207, 11.6%; Fishers exact p=0.00005; Extended Table 1). Similarly, delineating high and low post-transfusion recipient antibody levels above the geometric mean of 540 anti-S-RBD IgG titer, although less precise than AUC, revealed that participants receiving early treatment with high titer CCP also had no hospitalizations (0/45, 0%; Extended Fig. 1 and Table 1).
Donor antibody level pre-transfusion benchmarks of protection that prevent hospitalization
To correlate matched CCP donor units in the unvaccinated recipients to hospital outcome, we next investigated if the corresponding donor antibody levels would also translate into protection from hospitalization. The unvaccinated CCP recipients were again delineated by symptom onset to transfusion, but instead of post-transfusion antibody level representation, the matched corresponding donor antibody levels were stratified by donor anti-S-RBD IgG antibodies at GM 3286 AUC for all 592 recipients (Fig. 3A) and virus neutralization antibody (nAb) levels ≥60 AUC (Fig. 3B). In all antibody level metrics, we compared the GM upper to lower as high and low antibody levels, respectively. The numbers remained statistically significant by the Fishers exact test compared to both control plasma groups (Fig. 3 and Extended Table 1). Similar to the analyses with AUC values, donor anti-S-RBD IgG and nAb GM titer values, although less precise, revealed their respective recipients with early treatment and high donor antibody titers having the lowest proportion of hospitalizations (Extended Fig. 2 and Table 2). We also stratified donor antibody levels by the commercial assay EUROIMMUN ratio of optical density expressed above or below 6 AU or 600 BAU/mL (Extended Fig 3 and Extended Table 1), a level that also demarcates the upper portion of study donors24.
Screen seropositive hospitalizations in the unvaccinated
The screen seropositive recipients with subsequent hospitalizations were investigated, despite low incidence. 7/54 unvaccinated participants, subsequently hospitalized, were seropositive at the initial screen, two in the CCP group and five in the control group (Extended Table 2). Three of the five hospitalized participants from the control group had screen anti-S-RBD IgG antibody titers at 540 and AUC less than 150, fitting the early/low seronegative data described above. In the unvaccinated CCP group, 2/107 (1.9%) who were screen seropositive were hospitalized versus 15/378 (4%) seronegative at screen (Chi square p=0.29; CCP seropositive versus seronegative). Among the control group of whom were hospitalized, there were 5/92 (5.4%) screen seropositive versus 29/381 (7.6%) screen seronegative (chi square p=0.47 control-seropositive to seronegative). Of those screened seropositive, there was no statistical difference in hospitalization between those who received CCP and control plasma (chi square p=0.17; CCP-seropositive to control-seropositive). The single screen seronegative and partially vaccinated hospitalized participant transfused with control plasma received their first vaccine dose the same day as symptom onset. Among the 159 fully vaccinated and not hospitalized, 158 (99%) were screen seropositive, while 30/58 (69%) partially vaccinated participants were seropositive.
Pre-transfusion nasal SARS-CoV-2 viral RNA loads
Nasal viral load might independently determine risk of hospitalization. Quantitative RT-PCR was performed on the screen nasal swabs collected before plasma transfusion to correlate the SARS-CoV-2 nasal viral RNA load to hospitalization. All unvaccinated individuals subsequently receiving either control plasma or CCP had indistinguishable screen viral loads (i.e., near 10,000 copies) (Fig. 4A). Analyzing nasal viral loads of early/high, early/low, late/high, and late/low recipient groups revealed, as expected, that early transfusions closer to symptom onset were associated with higher viral loads than late transfusions. The CCP early/high and early/low recipient pre-transfusion nasal viral loads were indistinguishable. The viral load was similar among unvaccinated at screen, regardless of subsequent hospitalization outcome (Fig. 4B).
While the inclusion criteria required a documented positive molecular SARS-CoV-2 test (87% by RNA detection and 13% by antigen detection), the interval between the study inclusion SARS-CoV-2 test (performed outside the study) and the research study nasal swab collection prior to transfusion may have been up to a week.
Stratifying nasal viral load by days from symptom onset to transfusion time among unvaccinated participants infected prior to the Delta variant, which has different viral load kinetics26,27, indicated a decline in pre-transfusion viral burden among seronegative individuals after 5 days from symptom onset (Fig. 4C). Seropositive individuals had lower viral loads compared to seronegative individuals at all timepoints, with a majority of seropositive viral loads below the limit of detection (330 copies) by day 4 post-symptom onset (Fig. 4D). The screen viral RNA copy data suggest that seropositivity at the time of transfusion correlated with lower viral loads, but differences in viral RNA load did not impact hospitalization outcome.
Differences in antibody levels weeks after transfusion
Passive transfer of SARS-CoV-2 specific antibodies was postulated to possibly down-modulate subsequent SARS-CoV-2 antibody responses. This clinical study was designed to measure antibody responses up to 90 days post-transfusion to compare antibody kinetics between CCP and control plasma recipients. There were no differences in antibody levels between CCP and control plasma recipients at or beyond 14 days post-transfusion (Fig. 5). Longitudinal differences in antibody levels observed were due to hospitalization rather than treatment. The multivariate linear mixed-effects regression, adjusted for variant, age, sex, and BMI, showed no differences in antibody levels between CCP and control plasma recipients beyond 14 days post-transfusion (Fig. 5). There were neither sex, age, BMI, nor comorbidity differences in antibody levels between CCP and control groups.
Within the control group, there were eight immunocompetent recipients with undetectable SARS-CoV-2 antibody levels during all follow-up visits. Three of the 8 had persistent COVID-19 symptoms after 90 days of study follow-up. All CCP recipients had detectable antibodies at or after the day 14 visit.
Variant period differences in screen seropositivity, nasal viral load and day 90 antibody levels
We investigated how SARS-CoV-2 variants influenced seropositivity, nasal viral load and 3-month antibody levels in the setting of ongoing vaccination after December 2020. We separated participants into three periods: pre-Alpha (June 3, 2020 to January 31, 2021), Alpha (February 1, 2021 to July 15 2021), and Delta (July 16, 2021 to October 1, 2021). Participants recruited in the Delta period were younger, with fewer medical conditions, and more than 60% fully vaccinated (Extended Table 3). Screen seropositivity rates among unvaccinated individuals were low (∼ 20%), during pre-Alpha and Alpha periods rising to 46% during the Delta period. The baseline screen anti-S-RBD AUC GM antibody levels in the unvaccinated seropositive participants were 529, 482, and 7042 anti-S-RBD AUC during the pre-Alpha, Alpha and Delta periods, respectively compared to the 3286 AUC in the mainly pre-Alpha study donors (Extended Fig. 4A).
Pre-transfusion nasal viral loads were similar between CCP and control during the pre-Alpha, Alpha, and Delta periods (Extended Fig. 4B). When unvaccinated participants were stratified based on seropositivity at the time of transfusion, viral loads were consistently lower in seropositive individuals compared to seronegative individuals regardless of the time period in which CCP was administered (Extended Fig. 4C). During the Delta period, unvaccinated seropositive individuals also had lower viral loads compared to seronegative individuals, but the participant numbers stratified by symptom onset day were low (Extended Fig. 5A, B). Fully vaccinated and seropositive participants at screen also showed a drop in viral load when transfusion occurred more than 5 days after symptom onset (Extended Fig. 5C).
Comparing CCP and control day 90 antibody levels in those unvaccinated at screen (excluding the 165 vaccinated during the follow-up visits), indicated no difference between treatment groups or groups stratified by early or high CCP antibody treatment (Extended Figure 6A).
Those unvaccinated participants, subsequently hospitalized (41/54, 76%) during the pre-Alpha period, had a GM of 13007 AUC with very few having antibody levels below the mean for unvaccinated participants in the pre-Alpha period at day 90 compared to the non-hospitalized unvaccinated pre-Alpha participants GM 2691 AUC antibody level, 5 times less than those hospitalized (Extended Fig. 6B). Unvaccinated recipients during the Alpha period had similar day 90 antibody levels (geometric mean AUC=6683) to those infected during the Delta period (geometric mean AUC=5929), but near double the pre-Alpha period participants. Fully vaccinated recipients at screen who had breakthrough infection during Delta period showed a GM AUC of 53813, which was 20 times greater than the GM AUC of day 90 pre-Alpha period recipients.
Discussion
This jointly stratified study analysis by both time to transfusion and antibody levels found that post-transfusion anti-S-RBD IgG AUC levels at or over 150 AUC and transfusion within 5 days of symptom onset, resulted in no hospitalization in unvaccinated recipients. In seronegative recipients, the post-transfusion upper 50% antibody levels were proportionately matched to upper 50% donor antibody levels measurements by three different test methodologies-anti-S-RBD IgG ELISA, direct live virus neutralization, and EUROIMMUN anti-SARS-CoV-2 ELISA. Given that we selected the upper 60% of donors for study qualification and by all antibody measurements for stratified analysis, the upper half of qualified donors which equals the upper 30%, had similar impact on hospitalization risk reduction. The four-quadrant graphic depiction of hospitalizations was similar in the recipient antibody levels compared to the 3 separate donor antibody level measurements. This implicates the upper 30% of all potential pre-Alpha CCP donors as effective for reducing hospitalizations prior to the Delta variant.
An individual’s circulating plasma volume is, on average, 3 liters, which predicts a 15-fold proportional antibody dilution for a 200 mL CCP transfusion. A prior infection prevention study transfused CCP into healthy nonSARS-CoV-2 infected individuals and similarly measured 20-fold SARS-CoV-2 antibody dilution28. This suggests that the antibody compartment volume is similar between non-infected individuals and those with COVID-19 during the first week of illness. We did not observe a measurably higher volume of distribution, suggesting that extravascular transfer of high levels of antibodies to lower the post-transfusion antibody levels occurred. The donor and recipient antibody levels are directly proportional in Fig. 1B-C where the lines are parallel with the 22-fold dilution. From the recipient post-transfusion levels, one can predict matched donor levels are proportionately greater by a factor of 20. The screen seronegative participants had a clean negative antibody background. Those who were screen seropositive in Fig. 1D already had greater antibody levels than more than half of the post-transfusion seronegative participants. We have not explored whether CCP has better neutralizing abilities than the early host immune response when antibodies are just starting to measurably increase after 4 days from symptom onset for screen seropositive participants (low antibodies in the 500 to 1000 range) (Fig. 1E).
Antivirals (antibodies or small molecule antivirals) administered early in the COVID-19 course protect from progression to severe disease requiring hospitalization. At least three factors interact to predict risk of outpatient hospitalization: 1) viral load and variant properties; 2) human host risk factors for severe disease (age, obesity, or medical comorbidities), and 3) quantity and quality (degree of match to variants) of SARS-CoV-2 specific antibodies at disease onset, due to active vaccination or immune status (i.e., hybrid immunity, breakthrough, etc.).
We found no differences in nasal viral loads between control and CCP recipients at screening to account for differences in hospitalization rates. Importantly, those participants who were subsequently hospitalized had similar viral loads at the time of transfusion when compared to those not hospitalized. Unvaccinated recipients who were seropositive at screening had lower viral loads than seronegative participants. We also observed lower viral loads in those screened late. Specifically, those who were screened and transfused late (≥5 days after symptom onset) had lower viral loads compared to those who were transfused early, suggesting that the administration of antiviral drugs or CCP early (i.e., during the period of higher viral loads measured by nasal sampling) maximizes the therapeutic effect. Viral load dynamics in the lung or other tissues may differ, and treatment timing should be explored further.
The jointly stratified subgroup analysis was matched for patients’ age, demographics, obesity and one or more medical conditions for severe COVID-19 progression to hospitalization. Our study population was also younger, less obese and with fewer risk factors for disease progression than other published RCTs of CCP outpatient use29. Our study population also had a higher prevalence of seronegative participants upon transfusion than the Emergency Department based study (C3PO) where the seropositive rate was near 50%, which may be indicative of a different patient population30.
In this study, there are COVID-19 participants with a 100-fold range of antibody levels. CCP units from the top 30% of all convalescent donors are necessary to confer protection against hospitalization with severe COVID-19, such that when antibody levels are diluted approximately 20-fold, the recipient antibody levels remain above the CCP GM. These data suggest that convalescent plasma has been underdosed in prior infectious disease outbreaks and that future pandemic patients should receive higher therapeutic donor antibody levels. The current study introduces the concept that therapeutic donor CCP levels must be in the upper half or higher to fully account for the dilution effect in the recipient. Plasma from approximately 330 unique CCP donations were transfused into 592 CCP participants. These donor units were previously characterized for full-length anti-Spike IgG with GM titers of 13,053 and more precise AUC GM of 7938 which equals 243 BAU/mL using the international standards31. The median nAb titer was 80 with GM titer of 58 and nAb AUC of 51, equaling GM 27 IU/mL. The commercial EUROIMMUN arbitrary units (AU) mean was 6 for the unique units. The donor anti-S-RBD IgG GM titer was 6,678 and anti-S-RBD IgG GM AUC of 3,172. These thresholds separate the upper and lower portion of donor antibody levels and was also approximately 50% of total full length Spike titer antibody levels, but 100 times the nAb GM titer levels.
Unvaccinated participants who received CCP after 5 days (late) or received units with low antibody levels still had near significant reduction in hospital risk of about 4% rather than 7.6% in all controls. The parent study included fully vaccinated and partially vaccinated participants in the analyses, which lowered risk of hospitalization to 6.3%. Here, we observed an 11.6% risk of hospitalization among unvaccinated controls transfused early within 5 days of symptom onset. The effect of early CCP transfusion and high post-transfusion antibody is even greater among this subgroup of unvaccinated recipients.
The antibody level that affords protection is not absolute, as not even monoclonal antibody or small molecule antiviral therapy affords 100% reduction in risk of hospitalization. There were two screen seropositive participants in the CCP group and two in the control group with antibody levels also above the anti-S-RBD IgG antibody threshold of inverse dilution titer over 540 and AUC over 150, presumptively with newly formed antibodies. The finding that early administration of antibodies to SARS-CoV-2 is beneficial in reducing progression of disease is consistent with the observation that those who mount early antibody responses have lower rates of death after hospitalization32,33 and that inducement of humoral immunity by vaccination reduces severity of disease and death34.
When CCP was first deployed in 2020, there were concerns that specific antibody administration to individuals in the early stages of COVID-19 could interfere with the development of endogenous immune responses35. However, our findings show that transfusion of CCP, as compared to control plasma, was not associated with differences in the development of a humoral immune response in recipients, reassuring for the immunological safety of CCP in humans. The C3PO convalescent plasma study also did not see an antibody level difference between CCP and saline infusions30,36.
While our study had predominately SARS-CoV-2 naïve recipients enrolled prior to the Omicron variant who were largely unvaccinated, the findings are applicable to immunocompromised patients today who lack SARS-CoV-2 antibodies. Another limitation is the low number of seronegative participants transfused within 5 days of symptom onset with post-transfusion above the geometric mean donor antibody levels in our study population (approximately 100 participants). The parent study was not powered to look at these stratified quadrants.
In summary, our results support and reconfirm the adage that for antibody therapy to be effective, sufficient amounts of pathogen specific antibody should be dosed early37. No hospitalizations were observed in those treated at ≤5 days of symptom onset with high titer CCP indicating that this is the optimal combination for effective CCP use. Early treatment alone is insufficient as hospitalizations were still observed in the group treated ≤5 days with lower titer units, necessitating both early treatment and adequate antibody dosing for optimal efficacy. Our results provide evidence for the best use of CCP. We advocate that CCP units used for therapy comprise the upper 30% of donor antibody levels. These levels should set the threshold for future therapeutic CCP. When humanity faces its next pandemic, there is a high likelihood that convalescent plasma will be used again until better specific therapies become available. Our data provide a roadmap for optimal early, high dose convalescent plasma deployment in such future emergencies.
Online Methods
Study Ethics
Johns Hopkins served as the single-IRB (sIRB). For the Center for American Indian Health sites, the protocol was also independently reviewed and approved by the Navajo Nation Health Human Research Review Board and the National Indian Health Service IRB. The protocol was also approved by the Department of Defense (DoD) Human Research Protection Office (HRPO).
An independent medical monitor who was unaware of the trial group assignments reviewed all serious adverse events, and an independent panel of three physicians who were unaware of the trial-group assignments adjudicated Covid 19 related hospitalizations and severity. An independent data and safety monitoring board provided interim safety and efficacy reviews. The trial was conducted in accordance with the principles of the Declaration of Helsinki, the Good Clinical Practice guidelines of the International Council for Harmonisation, and all applicable regulatory requirements.
Study Population
In this multicenter, double-blind, randomized, controlled trial, we evaluated the efficacy and safety of COVID-19 convalescent plasma, as compared with control plasma, in symptomatic adults (≥18 years of age) who had tested positive for severe acute respiratory syndrome coronavirus 2, regardless of their risk factors for disease progression or vaccination status.
Participants were enrolled within 8 days after symptom onset and received a transfusion within 1 day after randomization. The primary study outcome (reported previously) was COVID-19– related hospitalization within 28 days after transfusion. There were no obvious imbalances between the trial groups in the parent trial with respect to baseline characteristics, including coexisting conditions, COVID-19 vaccination status, vital signs, and clinical laboratory results.
Study Center(s)
Anne Arundel Medical Center; Ascada Research; Baylor College of Medicine; Johns Hopkins Center for American Indian Health; Johns Hopkins Bloomberg School of Public Health; Johns Hopkins University; Lifespan/Brown University Rhode Island Hospital; Mayo Clinic, Phoenix; MedStar Washington Hospital Center; NorthShore University Health System; The Bliss Group; The Next Practice Group; University of California, Los Angeles Health; University of Alabama at Birmingham; University of California, Irvine Health; University of California, San Diego; University of Cincinnati Medical Center; University of Massachusetts Worcester; University of Miami; University of New Mexico; University of Rochester; University of Texas Health Science Center at Houston; University of Utah Health; Vassar Brothers Medical Center; Wayne State University; Western Connecticut Health Network, Danbury Hospital; Western Connecticut Health Network, Norwalk Hospital.
Study Plasma
The study qualified donor plasma with SARS-CoV-2 positive antibodies after a 1:320 dilution under FDA IND 19725 protocol. After July 2021, the transfused plasma donor units met the existing FDA Emergency Use Authorization (EUA) criteria for high titer at EUROIMMUN arbitrary unit (AU) over 3.5. Many identical apheresis donor plasma units were transfused into 2, 3, or 4 separate recipients. Plasma from 333 unique CCP donations was transfused into the 592 CCP participants. Seventy-five percent of the donor collections were before September 2020 with more than 90% by January 2021 and the last 25 collections by March 2021. These donor units were previously characterized for full-length anti-Spike IgG geometric mean (GM) titers of 13,053, which corresponded with a more precise area under the curve (AUC) geometric mean of 7938, equaling 243 BAU/mL using the international standards24. The median neutralizing antibody (nAb) titer was 80, with a geometric mean titer of 58, and nAb AUC of 51, equaling GM 27 IU/mL24. The commercial EUROIMMUN arbitrary units (AU) mean was 6 for the unique donor units24.
Study visits and time periods
In these studies, antibody levels were measured at screen before transfusion, within 30 minutes of transfusion, and various timepoints up to 90 days post-transfusion. Participants were transfused during pre-Alpha (June 3, 2020 to January 31, 2021), Alpha (February 1, 2021 to July 14, 2021), and Delta (July 15 to October 1, 2021) variant periods. There were just three participants transfused from July 2 to July 9, 2021 which decreased the number of false designations. The first Alpha (B1.1.7) confirmed by sequencing was from a participant transfused February 18, 2021.
EUROIMMUN ELISA Assay
The EUROIMMUN anti–SARS– CoV-2 ELISA for IgG (cat. EI2606-9601G) was validated in a Clinical Laboratory Improvement Amendments–certified (CLIA-certified) laboratory for donor qualification as positive after a 1:320 dilution as per IND 19725 protocol. The assay was performed according to the manufacturer’s specifications. In a separate research laboratory, the donor optical density (OD) was measured at 1:101 dilution. The ratio calculated by dividing the sample OD by the OD of the calibrator from that run constitutes the AU.To measure anti–SARS– CoV-2 IgG binding, each plate had the following components: 100 μL plasma (1:101 dilution, per the manufacturer’s protocol) and 100 μL undiluted positive, negative, and calibrator controls. Plates were washed 3 times, followed by the manufacturer’s protocol for addition of conjugate and substrate. Ratios of 0.8 or higher were considered positive.
Indirect ELISA
The ELISA protocol was adapted from a protocol published by the Florian Krammer laboratory38.The 96-well plates (Immulon 4HBX, Thermo Fisher Scientific-Cat#-3855) were coated with S-RBD of the parent strain at a volume of 50 μL of 2 μg/mL diluted antigen in filtered, sterile 1 × PBS (Thermo Fisher Scientific) at 4°C overnight. The coating buffer was removed, and the plates were washed 3 times with 300 μL 1 × PBS plus 0.1% Tween-20 (PBST) wash buffer (Thermo Fisher Scientific) and then blocked with 200 μL PBST with 3% nonfat milk (milk powder, American Bio) by volume for 1 hour at room temperature. All plasma samples were heat-inactivated at 56°C on a heating block for 1 hour before use and diluted 1:2 in PBS. Negative control samples were prepared at 1:10 dilutions in PBST in 1% nonfat milk and plated at a final dilution of 1:100. A mAb against the SARS-CoV-2 Spike protein was used as a positive control (1:5000 dilution; Sino Biological, 40150-D001). Plasma samples were prepared in 3-fold serial dilutions starting at 1:20 in PBST in 1% nonfat milk. Blocking solution was removed, and 100 μL diluted plasma was added in duplicate to the plates and incubated at room temperature for 2 hours. Plates were washed 3 times with PBST wash buffer, and 50 μL of secondary antibody was added to the plates and incubated at room temperature for 1 hour.
Antihuman secondary antibody, Fc-specific total IgG HRP (1:5000 dilution; Thermo Fisher Scientific, Invitrogen, A18823), was prepared in PBST plus 1% nonfat milk. Plates were washed, and all residual liquid was removed before the addition of 100 μL SIGMAFAST OPD (o-phenylenediamine dihydrochloride) solution (MilliporeSigma) to each well, followed by incubation in darkness at room temperature for 10 minutes. To stop the reaction, 50 μL 3M HCl (Thermo Fisher Scientific) was added to each well. The OD of each plate was read at 490 nm (OD490) on a SpectraMax i3 ELISA Plate Reader (BioTek Instruments). The positive cutoff value for each plate was calculated by summing the average of the negative values and 3 times the SD of the negatives. Limits of detection (LOD) were set to half the lowest AUC value at or below 20 titer. The anti-S-RBD IgG titer threshold for seronegative was 180 titer or below. The seropositive anti-S-RBD IgG ELISA titers represent 3-fold dilutions from 540 to 393,660.
SARS-CoV-2 Viral Load
Nasopharyngeal specimens obtained at screen were stored in 5 mL of virus transport media at - 70°C on site, then shipped to the central storage facility at Johns Hopkins University. RNA was extracted from 200 μL transport media with either the Qiagen viral RNA extraction kit (Qiagen, Hilden, Germany), or the chemagic Viral RNA/DNA 300 H96 kit with chemagic 360 nucleic acid extraction system (Perkin Elmer), according to manufacturer recommended protocols. Real-time reverse transcriptase quantitative PCR (RT-qPCR) assays targeting the SARS-CoV2 nucleocapsid (N) gene and the human RNaseP gene were performed based on the methods described by the US CDC39. Primer and FAM-labelled probe sets for CDC nCoV_N1 and RNaseP assays were purchased from IDT (Integrated DNA Technologies) as part of the SARS-CoV2 Research Use Only RUO qPCR primer and probe kit (part number 10006713, 2019_nCoV RUO kit). Single-plex assays with equivalent volumes of RNA (or Positive Control, Plasmid-RNA Standards or Nuclease Free H2O for No Template Controls (NTCs)) were performed using the TaqPath 1-Step RT-qPCR MasterMix (Applied Biosystems, ThermoFisher Scientific) in a QuantStudio 5 Real-Time PCR system (ThermoFisher Scientific). The SARS-CoV-2 nCoV-N control plasmid comprised the complete nucleocapsid gene of SARS-CoV-2 isolate Wuhan-Hu-1, complete genome (GenBank: NC_045512.2), and the HsRPP30 Positive control contained a portion of the RNAseP (RPP30) gene. Both plasmid controls were purchased from IDT. Standards for quantitative analysis were prepared from serial dilutions of the nCoV-N and HsRPP30 plasmid controls for which target copy number was known. The range covered was 200,000 copies to 320 copies. Standard curve analysis of nCoV_N1 Ct values was performed by the QuantStudio Design and Analysis software to determine RNA copies of viral genome. Only samples with quantities within the standard curve range were given a COVID-19 call/score “positive”. A Ct value for the RNaseP gene was used to verify that human RNA was present in each specimen. For samples that did not amplify viral genome or any host cell RNA, a repeat RT-qPCR was performed and subsequently assigned as “undetermined”.
SARS-CoV-2 Virus Neutralization Assay
Plasma neutralizing antibodies were determined against WA-1 (SARS-CoV-2/USA-WA1/2020 EPI_ISL_404895), which was obtained from BEI Resources, as described previously22,40. Two-fold dilutions of plasma (starting at a 1:20 dilution) were made and infectious virus was added to the plasma dilutions at a final concentration of 1 × 105 TCID50/mL (100 TCID50 per 100 μL). The samples were incubated with the virus for 1 hour at room temperature, and then 100 μL of each dilution was added to 1 well of a 96-well plate of VeroE6-TMPRSS2 cells in hexaplicate. The cells were incubated for 6 hours at 37°C, 5% CO2. The inocula were removed, fresh infection media (IM) was added, and the cells were incubated at 37°C, 5% CO2 for 2 days. The cells were fixed by the addition of 100 μL of 4% formaldehyde per well, incubated for at least 4 hours at room temperature, and then stained with Napthol Blue Black (MilliporeSigma). The neutralizing antibodies titer was calculated as the highest serum dilution that eliminated the cytopathic effect in 50% of the wells (NT50), and the AUC was calculated using Graphpad Prism.
Statistical analysis
Spearman correlation was used to calculated strength of association between titer and AUC units for antibody measurement. Fold drops in anti-S-RBD IgG AUC between donor and corresponding recipients were calculated by dividing the geometric mean of donors by that of recipients. Statistical differences between donor and post-transfusion recipient antibody levels were determined by non-parametric Mann-Whitney tests. Using viral load or antibody data, multiple comparisons across vaccination status, variant, serostatus, or treatment groups were performed using non-parametric Kruskall-Wallis tests with Dunn’s post-hoc corrections. Fishers exact tests were performed to compare the association of timing of treatment (early or late) and antibody levels (high or low) with hospitalization using unvaccinated, CCP recipient post-transfusion (D0) antibody data. Longitudinal recipient antibody data were first log10-transformed and analyzed using a linear mixed-effects regression model, adjusted for variant, age, sex, and BMI. An interaction term was included to examine how antibody levels changed over time by treatment (control or CCP) and hospitalization status for unvaccinated participants. Marginal effects were graphed with 95% confidence intervals. Statistical analyses were performed using GraphPad Prism 8 (GraphPad Software) and Stata 17 (StataCorp).
Data Availability
All data produced in the present study are available upon reasonable request to the authors
Data Availability
Source data for figures included in supplementary source data file. Data is available from individual authors upon request with reply expected in 14 days. Deidentified data from clinical trial will be deposited in the Vivli server for public access before end of 2023.
Code Availability
Unique software or computational code was not created for this study.
Author Contributions
K.L., N.M., D.E.F, L.J.A., B.L., D.H., A.C., S.S., E.B., K.G., A.T., O.L., A.P., S.L.K., D.J.S conceived and/or designed the work. HS.P., C.B., A.Yin, J.L., C.C., Y.E., R.F., O.B., J.S., T.G., P.C., J.W., N.N.,M.B., A.T., O.L., S.L.K., D.J.S contributed to the experimental design and procedures for the anti-S-RBD antibody level measurement. M.L., S.Y., I.S., A.J., A.P. contributed to the experimental design and procedure for the viral load and neutralizing antibody measurements. A.G.S., G.M.,Y.F., B.F., S.H., A.L., B.M., E.S., S.A., M.H., J.B., J.C., J.H.P., J.G., J.P., P.B, W.R., ME.C, J.H., B.G., V.C., D.C., K.O., M.A., L.H., C.S., D.N.F., M.Z., E.C., J.R., S.G.K., C.M., M.R., A.Yarava, K.L., N.M., A.G., N.K., A.S., D.E.F, D.A.J., L.J.A., D.M.S., B.L., S.E., S.B., T.G., A.Z., D.H., A.C., S.S., E.B., K.G., A.T., O.L., A.P., S.L.K., D.J.S conducted/contributed to the clinical study and/or collected clinical data. HS.P., C.B., A.Yin, J.L., C.C., M.L., S.Y., I.S., A.J., M.R., R.F., O.B., J.S., J.O., T.G., P.C., D.H., A.C., K.G., A.T., O.L., A.P., S.L.K., D.J.S contributed to data processing and analyses specific to this work. HS.P., C.B., A.Yin, A.C., A.P., S.L.K., D.J.S drafted the manuscript. All authors provided final approval of the version to be published.
Competing Interests
TG-Paid consultant and employee of Fenwal, a Fresenius Kabi company; AC-Scientific Advisory Board of Sabtherapeutics (cow-derived human immunoglobulins COVID-19 treatment and other infectious diseases) and Ortho Diagnostics Speakers Bureau; EB-member of the FDA Blood Products Advisory Committee; SS reports research grants; F2G, Cidara, Ansun, Zeteo, Emergent Biosolutions: personal fees as consultant, advisory board, data safety monitoring board member; Celltrion, Adagio, Immunome, Adamis, Karyopharm, Intermountain Health: Stock options: Immunome; CS: Centers for Disease Control and Prevention, Merck, Pfizer: Research Grants. All other authors report no relevant disclosures.
Tables
Extended Figures
Acknowledgements
Supported by a contract (W911QY2090012, to Dr. Sullivan) with the Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense of the Department of Defense, in collaboration with the Defense Health Agency; Bloomberg Philanthropies; the State of Maryland; a grant (3R01AI152078-01S1, to Dr. Casadevall) from the National Institutes of Health (NIH) National Institute of Allergy and Infectious Diseases (NIAID); a grant (U24TR001609-S3, to Dr. Hanley) from the NIH National Center for Advancing Translational Sciences; a grant (1K23HL151826NIH, to Dr. Bloch) from the National Heart, Lung, and Blood Institute; the Division of Intramural Research, NIAID, NIH; the Mental Wellness Foundation; the Moriah Fund; Octapharma; the Healthnetwork Foundation; and the Shear Family Foundation. The study sponsors did not contribute to the study design, the collection, analysis, and interpretation of data, and the decision to submit the paper for publication. We thank all the trial participants who generously gave of their time and donated biologic specimens. We thank Dr. Geeta Sood for a critical review of the manuscript.
Footnotes
These authors jointly supervised this work: Sabra Klein, David Sullivan
↵* David Sullivan can be contacted at dsulliv7{at}jhmi.edu or Sabra Klein at sklein2{at}jhu.edu; Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, 615 N. Wolfe St., Baltimore, MD 21205.
Abbreviations
- CCP
- COVID-19 convalescent plasma
- RCT
- randomized controlled trial