ArticlePDF Available

Passive immunotherapies for COVID‐19: The subtle line between standard and hyperimmune immunoglobulins is getting invisible

Authors:
DOI: 10.1002/rmv.2341
EDITORIAL
Passive immunotherapies for COVID19: The subtle line
between standard and hyperimmune immunoglobulins is
getting invisible
During the COVID19 pandemic, immunoglobulins (IG) have been
suggested both as an immunosuppressive treatment in late COVID
19 stages and as an antiviral in preexposure prophylaxis, post
exposure prophylaxis, and treatment of early COVID19 stages. As
immunosuppressants in late COVID19 stages, we and other groups
were not able to detect any signal of efficacy.
1
On the antiviral side, human neutralising IG (hyperimmune IG,
HIG) has been traditionally derived from selected convalescent plasma
(CP) donors.
2
The Inpatient Treatment of COVID19 With Anti
Coronavirus Immunoglobulin (ITAC) randomized controlled trial
[NCT04546581] of intravenous COVIDHIG (NP028) (0.4 g/kg) for
treatment of COVID19 inpatients (N=593) showed no benefit,
3
but
trials in outpatients are ongoing (INSIGHT 012, NCT04910269). To
date, COVIDHIG (NP028) has been shown to retain neutralising
potency against past variant of concern/interest Alpha, Beta, Gamma,
Delta/Delta+, Eta, Iota, Kappa, Lambda, Mu, but no data are available
for Omicron.
4
Furthermore, hyperimmune sera can nowadays be
manufactured from naïve vaccinees: for example, RojasJimenez et al.
used caprylic acid precipitation to prepare a pilotscale batch of anti
SARSCoV2 HIG from plasma of donors immunised with the
BNT162b2 (PfizerBioNTech) antiCOVID19 vaccine, showing higher
concentration of antiRBD and ACE2RBD neutralizing antibodies
(nAbs) than in CCPderived IVIg.
5
But HIG manufacturing suffers from
logistical bottlenecks, which hamper scalability. The availability of
nAbs within a product which is already scaled up would facilitate large
scale deployment.
Despite initial signs of heterologous immunity to SARSCoV2
from previous seasonal coronavirus infection in prepandemic IG
lots of HyQvia
®
(Baxalta Innovations GmbH), Privigen
®
(CSL Behr-
ing), Intratect
®
(Biotest AG), IgVena
®
(Kedrion S.p.A), and Flebo-
gamma
®
(Grifols S.A.),
6–8
prepandemic sera have been shown to be
devoid of nAb,
9,10
making the occurrence of neutralizing activity
extremely unlikely in IG lots manufactured from plasma collected
before 2021. However, the situation is rapidly evolving, with most
plasma donors worldwide becoming SARSCoV2 seropositive
because of convalescence and/or COVID19 vaccination.
Karbiener et al. at Baxter reported minimal seropositivity in IG
lots released since September 2020: from there, values steadily
increased, in correlation with the cumulative COVID19 incidence, to
reach a mean of 36.7 international units (IU)/ml, and 93% of IG lots
were positive by January 2021.
11
Extrapolating the correlation, the
authors estimated that IVIGs could have reached an antiSARSCoV
2 potency of 400 IU/ml (i.e., a dose similar to that contained in a
COVID19 convalescent plasma (CCP) unit) by July 2021.
11
Farcet et al. at Takeda tested 176 IG lots released since March
2020 for SARSCoV2 nAbs, with first positive results for September
2020 lots, mean =1.7 IU/ml, 46% of lots positive. From there, values
steadily increased, in correlation with the cumulative COVID19
incidence, to reach a mean of 31.2 IU/ml and 93% of lots positive
by January 2021. Extrapolating the correlation, intravenous immu-
noglobulins (IVIG) could reach an antiSARSCoV2 potency of 345
IU/ml by July 2021. At that stage, prophylactic IVIG treatment for
primary/secondary immunodeficiency could contain similar doses of
antiSARSCoV2 as CCP.
12
Volk et al. investigated SARSCoV2 reactivity of US plasma
derived IVIG/subcutaneous IG products from the end of 2020
until June 2021 as well as in CP from May 2020 to August 2020
to determine whether potentially neutralizing antibody titers may
be present.
13
While approximately 50% of convalescent donations
were not/low neutralizing, approximately 10% were at or above
600 IU/ml. Lots produced between December 2020 and June 2021
entailing plasma donations after the emergence of SARSCoV2
showed a rapid and constant increase in antiSARSCoV2 reac-
tivity and nAbs over time. While lottolot variability was sub-
stantial, neutralization capacity increased from a mean of 21 IU/ml
in December 2020 to 506 IU/ml in June 2021 with a maximum of
864 IU/ml for the most recent lots. Pharmacokinetic extrapola-
tions, based on noncompartmental superposition principles using
steadystate reference profiles from previously published pharma-
cokinetic investigations on IVIG in primary immunodeficiency,
yielded potential steadystate trough plasma levels of 16 IU/ml of
neutralizing SARSCoV2 IgG based on the average final container
concentration from May 2021 of 216 IU/ml. Maximum extrapo-
lated trough levels could reach 64 IU/ml based on the latest
maximal final container potency tested in June 2021.
13
Stinca et al. predicted that antiSARSCoV2 IgG concentration
will peak in batches produced in midOctober 2021, containing levels
in the vicinity of 190fold that of the mean (unvaccinated) CCP.
14
An
elevated concentration (approximately 35fold CCP) is anticipated to
be retained in batches produced well into 2022. Measurement of
Rev Med Virol. 2022;e2341. wileyonlinelibrary.com/journal/rmv © 2022 John Wiley & Sons Ltd.
-
1 of 3
https://doi.org/10.1002/rmv.2341
several Privigen batches using the Phadia™ EliA™ SARSCoV2Sp1
IgG binding assay confirmed the early phase of this model.
14
CP and IG (either regular IG or HIG) have different
manufacturing times, which makes CP preferrable early in pan-
demics, while IG take several months to be available.
3
Regardless
of indication (which is not expected at its best against a respira-
tory pathogen with marginal systemic spread), the 2 antiviral for-
mulations also differ in composition, with purified IG devoid of IgM
and IgA classes.
In conclusion, there are encouraging signals that regular IG will
equate HIG in terms of efficacy, while granting higher scalability and
lower costs. With fastwaning immunity (after either vaccination or
natural infections), it is not currently clear how long this opportunity
will last. Until this occurs, it is more clear which population subgroups
will most benefit from it. In particular, such IG should compete with
antiSpike monolonal antibodies (mAbs) for preexposure prophylaxis
in immunocompromised subjects who did not respond to vaccines,
but also for postexposure prophylaxis and early treatment in frail
subjects. This is becoming more and more relevant in sight of resis-
tance of Omicron to mAbs (including sotrovimabresistant BA.2
15
),
while, in contrast, CP from vaccinees is getting higher and higher
titers of nAbs.
16
The results of clinical trials are awaited with interest.
(Figure 1)
KEYWORDS
COVID19, immunosuppressants, intravenous immunoglobulins,
polyclonal antibodies, SARSCoV2
CONFLICT OF INTEREST
We declare we have no conflict of interest related to this manuscript.
AUTHOR CONTRIBUTIONS
D.F. conceived the manuscript and wrote the first draft. M.F. revised
the manuscript.
Daniele Focosi
1
Massimo Franchini
2
1
NorthWestern Tuscany Blood Bank, Pisa University Hospital,
Pisa, Italy
2
Department of Hematology and Transfusion Medicine, Carlo
Poma Hospital, Mantua, Italy
Correspondence
Daniele Focosi, NorthWestern Tuscany Blood Bank, Pisa University
Hospital, 56124 Pisa, Italy.
Email: daniele.focosi@gmail.com
REFERENCES
1. Focosi D, Franchini M, Tuccori M, Cruciani M. Efficacy of highdose
polyclonal intravenous immunoglobulin in COVID19: a systematic
review. Vaccines (Basel). 2022;10:94. https://doi.org/10.3390/
vaccines10010094
2. Focosi D, Franchini M, Tuccori M. The road towards polyclonal anti
SARSCoV2 immunoglobulins (hyperimmune serum) for passive
immunization in COVID19. Life (Basel). 2021;11:144.
3. Focosi D, Tuccori M, Antonelli G, Maggi F. What is the optimal usage
of Covid19 convalescent plasma donations? Clin Microbiol Infect.
2020;27(2):163165.
4. Toth D. Neutralization Of SARSCoV2 variants by a human poly-
clonal antibody therapeutic (COVIDHIG, NP028) with high
neutralizing titers to SARSCoV2. J bioRxiv. 2022:2027478053.
https://doi.org/10.1101/2022.01.27.478053
5. RojasJiménez G, Solano D, Segura Á, et al. In vitro characterization
of antiSARSCoV2 intravenous immunoglobulins (IVIg) produced
from plasma of donors immunized with the BNT162b2 vaccine and
its comparison with a similar formulation produced from plasma of
COVID19 convalescent donors. Front Med Technol. 2021;3:772275.
https://doi.org/10.3389/fmedt.2021.772275
6. Ng KW, Faulkner N, Cornish GH, et al. Preexisting and de novo
humoral immunity to SARSCoV2 in humans. Science.
2020;370:13391343. https://doi.org/10.1126/science.abe1107
7. Díez JM, Romero C, Gajardo R. Currently available intravenous
immunoglobulin contains antibodies reacting against severe acute
FIGURE 1 The natural evolution of neutralising antibodybased therapies against an emerging pathogen. VOC, variant of concern
2 of 3
-
EDITORIAL
respiratory syndrome coronavirus 2 antigens. Immunotherapy.
2020;12:571576. https://doi.org/10.2217/imt20200095
8. Dalakas MC, Bitzogli K, Alexopoulos H. AntiSARSCoV2 antibodies
within IVIg preparations: crossreactivities with seasonal coronavi-
ruses, natural autoimmunity, and therapeutic implications. Front
Immunol. 2021;12:627285. https://doi.org/10.3389/fimmu.2021.
627285
9. Focosi D, Rosellini A, Spezia PG, et al. Lack of neutralizing activity in
nonconvalescent sera, regardless of ABO blood group antiA iso-
agglutin titer. J Clin Virol Plus. 2021;1(3).
10. KubotaKoketsu R, Terada Y, Yunoki M, et al. Neutralizing and
binding activities against SARSCoV 1/2, MERSCoV , and human
coronaviruses 229E and OC43 by normal human intravenous
immunoglobulin derived from healthy donors in Japan. Transfusion.
2020;61:356360. https://doi.org/10.1111/trf.16161
11. Karbiener M, Farcet MR, Schwaiger J, et al. Plasma from post
COVID19 and COVID19vaccinated donors results in
highly potent SARSCoV2 neutralization by intravenous
immunoglobulins. J Infect Dis. 2021. https://doi.org/10.1093/infdis/
jiab482
12. Farcet MR, Karbiener M, Schwaiger J, Ilk R, Kreil TR. Rapidly
increasing SARSCoV2 neutralization by intravenous immunoglob-
ulins produced from plasma collected during the 2020 pandemic. J
Infect Dis. 2021. https://doi.org/10.1093/infdis/jiab142
13. Volk A, CoviniSouris C, Kuehnel D, De Mey C, Römisch J, Schmidt T.
SARSCoV2 neutralization in convalescent plasma and commercial
lots of plasmaderived immunoglobulin. BioDrugs. 2021;36:4153.
https://doi.org/10.1007/s40259021005119
14. Stinca S, Barnes TW, Vogel P, et al. Modelling the concentration of
antiSARSCoV2 immunoglobulin G in intravenous immunoglobulin
product batches. PLoS ONE. 2021;16:e0259731. https://doi.org/10.
1371/journal.pone.0259731
15. Iketani S, Liu L, Guo Y, et al. Antibody evasion properties of SARS
CoV2 omicron sublineages. J bioRxiv. 2022:479306. https://doi.
org/10.1101/2022.02.07.479306
16. Focosi D, Franchini M, Joyner MJ, Casadevall A. Comparative analysis
of antibody responses from COVID19 convalescents receiving
various vaccines reveals consistent high neutralizing activity for
SARSCoV2 variant of concern omicron. 2021:2021202421268317.
https://doi.org/10.1101/2021.12.24.21268317
EDITORIAL
-
3 of 3
... In the current post-vaccine era, however, the hybrid (i.e., vaccine and infection) exposure of large part of the population has led to high-titer heterologous immunity against SARS-CoV-2 providing an efficient cross-protection against most variants, including Omicron sub-lineages [74]. Within this new context, currently available standard IVIg, originating mostly from CCP donations, equate hIVIg in term of anti-SARS-CoV-2 nAb content, thus rendering the creation of a dedicated manufacturing chain poorly cost-effective and obsolete [75]. ...
Preprint
Full-text available
Since late 2019, COVID-19, a severe acute respiratory syndrome pandemic, was caused by the new SARS-CoV-2 virus belonging to the Coronaviridae family, . Several antiviral therapies, mostly derived from previous epidemics, were initially repurposed to fight this not rarely life-threatening respiratory illness. Among them, however, the only specific antibody-based therapy available against SARS-CoV-2 infection during the first year of the pandemic was represented by COVID-19 convalescent plasma (CCP). CCP, collected from recovered individuals, contains high levels of polyclonal antibodies of different subclasses able to neutralize SARS-CoV-2 infection. Tens of randomized controlled trials have been conducted during the last three years of the pandemic to evaluate the safety and the clinical efficacy of CCP in both hospitalized and ambulatory COVID-19 patients, whose main results will be summarized in this narrative review. In addition, we will present the current knowledge on the development of anti-SARS-CoV-2 hyperimmune polyclonal immunoglobulins.
... Further RCTs are needed to assess CCP in combination regimens. It remains to be established whether, at the steady state of viral evolution, concentrated polyclonal IgG formulations would be as equally effective as CCP [41,42]. ...
Article
Full-text available
COVID-19 convalescent plasma (CCP) has represented the frontline response to the COVID-19 pandemic, largely because of encouraging historical evidences in previous pandemics, biological plausibility, and the initial unavailability of targeted antivirals. Unfortunately, investigator-initiated randomized clinical trials in 2020, launched during a stressful pandemic peak, were designed mostly at addressing the main unmet need, i.e., treating critically ill hospitalized patients who were unlikely to benefit from any antiviral therapy. The failure of most of these drugs, in combination with the lack of any sponsor, led to the false belief that convalescent plasma was useless. With the relaxing pandemic stages, evidences have instead mounted that, when administered properly (i.e., within 5 days from onset of symptoms and at high titers of neutralizing antibodies), CCP is as effective as other antivirals at preventing disease progression in outpatients, and also reduces mortality in hospitalized patients. Recently, the focus of clinical use has been on immunosuppressed patients with persistent seronegativity and infection, where a randomized clinical trial has shown a reduction in mortality. Lessons learnt during the COVID-19 pandemic will be of utmost importance for future pandemics.
Chapter
Pandemics are highly unpredictable events that are generally caused by novel viruses. There is a high likelihood that such novel pathogens belong to entirely novel viral families for which no targeted small-molecule antivirals exist. In addition, small-molecule antivirals often have pharmacokinetic properties that make them contraindicated for the frail patients who are often the most susceptible to a novel virus. Passive immunotherapies—available from the first convalescent patients—can then play a key role in controlling pandemics. Convalescent plasma is immediately available, but if manufacturers have fast platforms to generate marketable drugs, other forms of passive antibody treatment can be produced. In this chapter, we will review the technological platforms for generating monoclonal antibodies and hyperimmune immunoglobulins, the current experience on their use for treatment of COVID-19, and the pipeline for pandemic candidates.
Article
Since the emergence of COVID-19 in 2020, an unprecedented range of therapeutic options has been studied and deployed. Healthcare providers have multiple treatment approaches to choose from, but efficacy of those approaches often remains controversial or compromised by viral evolution. Uncertainties still persist regarding the best therapies for high-risk patients, and the drug pipeline is suffering fatigue and shortage of funding. In this article, we review the antiviral activity, mechanism of action, pharmacokinetics, and safety of COVID-19 antiviral therapies. Additionally, we summarize the evidence from randomized controlled trials on efficacy and safety of the various COVID-19 antivirals and discuss unmet needs which should be addressed.
Article
Influenzavirus is among the most relevant candidates for a next pandemic. We review here the phylogeny of former influenza pandemics, and discuss candidate lineages. After briefly reviewing the other existing antiviral options, we discuss in detail the evidences supporting the efficacy of passive immunotherapies against influenzavirus, with a focus on convalescent plasma.
Article
The epidemic of corona virus disease 2019 (COVID-19) caused by severe acute respiratory syndrome Coronavirus 2 and its variants of concern (VOCs) has been ongoing for over 3 years. Antibody therapies encompassing convalescent plasma, hyperimmunoglobulin, and neutralizing monoclonal antibodies (mAbs) applied in passive immunotherapy have yielded positive outcomes and played a crucial role in the early COVID-19 treatment. In this review, the development path, action mechanism, clinical research results, challenges, and safety profile associated with the use of COVID-19 convalescent plasma, hyperimmunoglobulin, and mAbs were summarized. In addition, the prospects of applying antibody therapy against VOCs was assessed, offering insights into the coping strategies for facing new infectious disease outbreaks.
Article
Background and objectives: COVID-19 convalescent plasma (CCP) has retained potency and clinical efficacy against SARS-CoV-2 and is currently of utmost value for seronegative immunocompromised patients. Since most of the effect is due to the vaccine boost of infection-elicited antibodies, there is a theoretical concern that the frequency of suitable donors is declining. Materials and methods: In this single-institution serosurvey, we screened 599 consecutive donors attending our area in two different seasons (300 in November 2022 and 299 in February 2023) using the Abbott Alinity® anti-Spike immunoglobulin G assay. Results: More than 80% of random donors qualify according to the FDA criteria for high-titre CCP (>4350 AU/mL), with a stable trend. Conclusion: Despite reduced anti-Spike vaccine boost deployment in the general population, we have shown here that high-titre CCP units are easier than ever to procure. This finding also has implications for the derivation of standard immunoglobulins, which are finally approaching the potency of hyperimmune serum and could soon represent an alternative to CCP.
Article
To the Editor—We congratulate Maor and colleagues for their unprecedented randomized clinical trial (RCT) comparing convalescent plasma (CP) with CP-derived immunoglobulin G (cIgG) in hospitalized coronavirus disease 2019 (COVID-19) patients [1], which represents one of the very few head-to-head RCTs done during the pandemic. cIgG is devoid of immunoglobulin A and immunoglobulin M, which have a prominent role in virus neutralization [2], although each has a shorter half-life than IgG. Consequently, their finding that CP and cIgG have comparable efficacy at day 14, but not day 28 [1], has a potential pharmacokinetic explanation, as neutralizing antibody provided by CP would be cleared faster. The CP used in their RCT was qualified based on high-throughput serology (Abbott anti-Spike IgG >1050 AU/mL), with the US Food and Drug Administration recommending at least >1280 AU/mL [3]. Despite both arms receiving 4 g total antibody, anti-spike IgG in the 2 CP units (0.42 million AU) was estimated to be 25% the anti–spike 1 IgG in cIgG (1.35 million AU), which could also explain the trends to greater efficacy at day 14 and superiority at day 28. The superiority of cIgG at day 14 provides additional evidence for the importance of high antibody dosing in COVID-19. CP has consistently been shown to be safe and effective when used in the first 5 days since onset of COVID-19 symptoms [4], so the authors’ findings that there was no difference in response between those treated before versus after 7 days since onset of symptoms [1] is likely due to low titers in CP or late treatment, since those requiring hospitalization in the first 7 days from symptom onset likely had a more severe disease course. Finally, a subgroup analysis for the 67 immunocompromised patients (27 in the cIgG arm and 40 in the CP arm) could be informative.
Article
COVID-19 convalescent plasma (CCP) has been explored as one of the treatment options for COVID-19. Results of many cohort studies and clinical trials have been recently published. At first glance, the results of the CCP studies appear to be inconsistent. However, it became clear that CCP is not beneficial if CCP with low anti-SARS-CoV-2 antibody concentrations is used, if it is administered late in advanced disease stages, and to patients who already mounted an antibody response against SARS-CoV-2 at the time of CCP transfusion. On the other hand, CCP may prevent progression to severe COVID-19 when very high-titer CCP is given early in vulnerable patients. Immune escape of new variants is a challenge for passive immunotherapy. While new variants of concern developed resistance to most clinically used monoclonal antibodies very rapidly, immune plasma from individuals immunized by both a natural SARS-CoV-2 infection and SARS-CoV-2 vaccination retained neutralizing activity against variants. This review briefly summarizes the evidence on CCP treatment to date and identifies further research needs. Ongoing research on passive immunotherapy is not only relevant for improving care for vulnerable patients in the ongoing SARS-CoV-2 pandemic, but even more as a model for passive immunotherapy in case of future pandemics with a newly evolving pathogen. Compared to other drugs, which must be newly developed in a pandemic (e.g., monoclonal antibodies, antiviral drugs), convalescent plasma is rapidly available, inexpensive to produce, and can be adaptive to viral evolution by selection of contemporary convalescent donors.
Article
Full-text available
The identification of the Omicron variant (B.1.1.529.1 or BA.1) of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) in Botswana in November 20211 immediately raised alarms due to the sheer number of mutations in the spike glycoprotein that could lead to striking antibody evasion. We2 and others3-6 recently reported results in this Journal confirming such a concern. Continuing surveillance of Omicron evolution has since revealed the rise in prevalence of two sublineages, BA.1 with an R346K mutation (BA.1+R346K, also known as BA.1.1) and B.1.1.529.2 (BA.2), with the latter containing 8 unique spike mutations while lacking 13 spike mutations found in BA.1. We therefore extended our studies to include antigenic characterization of these new sublineages. Polyclonal sera from patients infected by wild-type SARS-CoV-2 or recipients of current mRNA vaccines showed a substantial loss in neutralizing activity against both BA.1+R346K and BA.2, with drops comparable to that already reported for BA.12,3,5,6. These findings indicate that these three sublineages of Omicron are antigenically equidistant from the wild-type SARS-CoV-2 and thus similarly threaten the efficacies of current vaccines. BA.2 also exhibited marked resistance to 17 of 19 neutralizing monoclonal antibodies tested, including S309 (sotrovimab)7, which had retained appreciable activity against BA.1 and BA.1+R346K2-4,6. This new finding shows that no authorized monoclonal antibody therapy could adequately cover all sublineages of the Omicron variant, except for the recently authorized LY-CoV1404 (bebtelovimab).
Preprint
Full-text available
Since the start of the COVID-19 outbreak the World Health Organization (WHO) has classified multiple SARS-CoV-2 Variants-of-Concern and Variants-of-Interest (VOC/VOI) with mutations in their Spike protein that increase transmissibility and/or reduce the effectiveness of vaccines and monoclonal antibody therapeutics. The emergence of these variants represents a significant health risk and highlights the need for additional COVID-19 therapeutics that maintain the ability to neutralize current, as well as future variants. COVID-HIG (NP-028) is a polyclonal Anti-SARS-CoV-2 human Immunoglobulin purified from source human plasma screened for high antibody titers to SARS-CoV-2 antigens. COVID-HIG was previously evaluated in INSIGHT 013 clinical trial [ NCT04546581 ] which was an international, multi-center, adaptive, randomized, double-blind, placebo-controlled trial of the safety, tolerability and efficacy of a single dose infusion (up to 400 mL) of Anti-Coronavirus Hyperimmune Intravenous Immunoglobulin (hIVIG) for the treatment of adult recently hospitalized COVID-19 patients (N=593). COVID-HIG is currently being evaluated for clinical efficacy in a Phase 3 placebo-controlled study INSIGHT 012 ( NCT04910269 ) to compare the safety and efficacy of a single infusion of anti-COVID-19 hyperimmune immunoglobulin (hIVIG) versus placebo among adults with recently diagnosed SARS-CoV-2 infection who do not require hospitalization. In the present study, in-vitro pseudovirus and live virus neutralization assays were used to assess the impact of SARS-CoV-2 variant spike mutations on neutralizing potency of COVID-HIG. These assays are valuable tools for monitoring the potential impact of variant mutations on efficacy of antibody therapeutics as well as vaccines/natural immunity. To date, COVID-HIG (NP-028) has been shown to retain neutralizing potency against 20 full spike protein sequence SARS-CoV-2 pseudovirus variants including all currently classified VOC/VOI (Alpha, Beta, Gamma, Delta/Delta+, Eta, Iota, Kappa, Lambda, Mu as of Sept 2021) as well as 4 live virus variants (Alpha, Beta, Gamma, and Iota).
Article
Full-text available
Background: Although several therapeutic strategies have been investigated, the optimal treatment approach for patients with coronavirus disease (COVID-19) remains to be elucidated. This systematic review and meta-analysis aimed to evaluate the efficacy and safety of polyclonal intravenous immunoglobulin (IVIG) therapy in COVID-19. Methods: A systematic literature search using appropriate medical subject heading (MeSH) terms was performed through Medline (PubMed), EMBASE, SCOPUS, OVID and Cochrane Library electronic databases. The main outcomes considered were mortality and safety of IVIG versus placebo/standard of care. This review was carried out in accordance with Cochrane methodology including the risk bias assessment and grading of the quality of evidence. Measures of treatment effect were mean differences (MD) together with 95% confidence intervals (CIs) for continuous outcome measures and risk ratio (RR) or MD for binary outcomes. Two reviewers independently extracted data from individual studies, and disagreements were resolved by a third reviewer. Results: A total of 2401 COVID-19 patients from 10 studies (four randomized controlled trials (RCT) and six non-randomized controlled trials (non-RCTs)) were included in the analysis. Participants received IVIG or placebo/standard of care. The use of IVIG was not associated with a significantly reduced risk of death (RR 0.50, 95% CIs 0.18-1.36, p = 0.17 for RCTs; RR 0.95, 95% CIs 0.61-1.58, p = 0.94 for non-RCTs; low certainty of evidence). IVIG significantly reduced the length of hospital stay (MD -2.24, 95% CIs -3.20/-1.27; p = 0.00001; low certainty of evidence), although this difference was significant only for studies evaluating moderate COVID-19 patients. No significant difference was observed in the incidence of overall and serious adverse events between IVIG recipients and controls (very low certainty of evidence). Conclusions: The current evidence from the literature does not support the use of IVIG in COVID-19 patients.
Article
Full-text available
Despite vaccines are the main strategy to control the ongoing global COVID-19 pandemic, their effectiveness could not be enough for individuals with immunosuppression. In these cases, as well as in patients with moderate/severe COVID-19, passive immunization with anti-SARS-CoV-2 immunoglobulins could be a therapeutic alternative. We used caprylic acid precipitation to prepare a pilot-scale batch of anti-SARS-CoV-2 intravenous immunoglobulins (IVIg) from plasma of donors immunized with the BNT162b2 (Pfizer-BioNTech) anti-COVID-19 vaccine (VP-IVIg) and compared their in vitro efficacy and safety with those of a similar formulation produced from plasma of COVID-19 convalescent donors (CP-IVIg). Both formulations showed immunological, physicochemical, biochemical, and microbiological characteristics that meet the specifications of IVIg formulations. Moreover, the concentration of anti-RBD and ACE2-RBD neutralizing antibodies was higher in VP-IVIg than in CP-IVIg. In concordance, plaque reduction neutralization tests showed inhibitory concentrations of 0.03–0.09 g/L in VP-IVIg and of 0.06–0.13 in CP-IVIg. Thus, VP-IVIg has in vitro efficacy and safety profiles that justify their evaluation as therapeutic alternative for clinical cases of COVID-19. Precipitation with caprylic acid could be a simple, feasible, and affordable alternative to produce formulations of anti-SARS-CoV-2 IVIg to be used therapeutically or prophylactically to confront the COVID-19 pandemic in middle and low-income countries.
Preprint
Full-text available
The novel SARS-CoV-2 Omicron variant of concern (VOCs), with its escape from unboosted vaccines and monoclonal antibodies, is demanding for a return to COVID19 convalescent plasma therapies. Lessons learnt from previous usage of CCP suggests focusing on outpatients and using high nAb-titer units in early disease stages. In this systematic analysis, we show that CCP from unvaccinated donors is not effective against Omicron, while CCP from vaccinees convalescents from previous VOCs or third-dose uninfected vaccinees is likely to remain effective against Omicron. countries. CCP remains the only antibody-based therapy that keeps up with the variants and provides an effective tool to combat the emergence of variants that defeat monoclonal antibodies. Consequently, there is a need for continue study of the variables that determine CCP efficacy.
Article
Full-text available
Background Plasma-derived intravenous immunoglobulin (IVIg) products contain a dynamic spectrum of immunoglobulin (Ig) G reactivities reflective of the donor population from which they are derived. We sought to model the concentration of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG which could be expected in future plasma pool and final-product batches of CSL Behring’s immunoglobulin product Privigen. Study design and methods Data was extracted from accessible databases, including the incidence of coronavirus disease 2019 and SARS-CoV-2 vaccination status, antibody titre in convalescent and vaccinated groups and antibody half-life. Together, these parameters were used to create an integrated mathematical model that could be used to predict anti-SARS-CoV-2 antibody levels in future IVIg preparations. Results We predict that anti-SARS-CoV-2 IgG concentration will peak in batches produced in mid-October 2021, containing levels in the vicinity of 190-fold that of the mean convalescent (unvaccinated) plasma concentration. An elevated concentration (approximately 35-fold convalescent plasma) is anticipated to be retained in batches produced well into 2022. Measurement of several Privigen batches using the Phadia ™ EliA ™ SARS-CoV-2-Sp1 IgG binding assay confirmed the early phase of this model. Conclusion The work presented in this paper may have important implications for physicians and patients who use Privigen for indicated diseases.
Article
Full-text available
Introduction: Patients with primary or secondary immunodeficiency (PID or SID) face increased insecurity and discomfort in the light of the COVID-19 pandemic, not knowing if and to what extent their comorbidities may impact the course of a potential SARS-CoV-2 infection. Furthermore, recently available vaccination options might not be amenable or effective for all patients in this heterogeneous population. Therefore, these patients often rely on passive immunization with plasma-derived, intravenous or subcutaneous immunoglobulin (IVIG/SCIG). Whether the ongoing COVID-19 pandemic and/or the progress in vaccination programs lead to increased and potentially protective titers in plasma-derived immunoglobulins (Ig) indicated (e.g., for humoral immunodeficiency) remains a pressing question for this patient population. Purpose: We investigated SARS-CoV-2 reactivity of US plasma-derived IVIG/SCIG products from the end of 2020 until June 2021 as well as in convalescent plasma (CP) from May 2020 to August 2020 to determine whether potentially neutralizing antibody titers may be present. Methods: Final containers of IVIG/SCIG and CP donations were analyzed by commercial ELISA for anti-SARS-CoV-2 S1-receptor binding domain (RBD) IgG as well as microneutralization assay using a patient-derived SARS-CoV-2 (D614G) isolate. Neutralization capacities of 313 single plasma donations and 119 plasma-derived IVIG/SCIG lots were determined. Results obtained from both analytical methods were normalized against the WHO International Standard. Finally, based on dense pharmacokinetic profiles of an IVIG preparation from previously published investigations, possible steady-state plasma levels of SARS-CoV-2 neutralization capacities were approximated based on currently measured anti-SARS-CoV-2 potencies in IVIG/SCIG preparations. Results: CP donations presented with high variability with regards to anti-SARS-CoV-2 reactivity in ELISA as well as in neutralization testing. While approximately 50% of convalescent donations were not/low neutralizing, approximately 10% were at or above 600 IU/mL. IVIG/SCIG lots derived from pre-pandemic plasma donations did not show neutralizing capacities for SARS-CoV-2. Lots produced between December 2020 and June 2021 entailing plasma donations after the emergence of SARS-CoV-2 showed a rapid and constant increase in anti-SARS-CoV-2 reactivity and neutralization capacity over time. While lot-to-lot variability was substantial, neutralization capacity increased from a mean of 21 IU/mL in December 2020 to 506 IU/mL in June 2021 with a maximum of 864 IU/mL for the most recent lots. Pharmacokinetic extrapolations, based on non-compartmental superposition principles using steady-state reference profiles from previously published pharmacokinetic investigations on IVIG in PID, yielded potential steady-state trough plasma levels of 16 IU/mL of neutralizing SARS-CoV-2 IgG based on the average final container concentration from May 2021 of 216 IU/mL. Maximum extrapolated trough levels could reach 64 IU/mL based on the latest maximal final container potency tested in June 2021. Conclusions: SARS-CoV-2 reactivity and neutralization capacity in IVIG/SCIG produced from US plasma rapidly and in part exponentially increased in the first half of 2021. The observed increase of final container potencies is likely trailing the serological status of the US donor population in terms of COVID-19 convalescence and vaccination by at least 5 months due to production lead times and should in principle continue at least until Fall 2021. In summary, the data support rapidly increasing levels of anti-SARS-CoV-2 antibodies in IVIG/SCIG products, implicating that a certain level of protection could be possible against COVID-19 for regularly substituted PID/SID patients. Nevertheless, more research is still needed to confirm which plasma levels are needed to provide protection against SARS-CoV-2 infection in immune-compromised patients.
Article
Full-text available
From September 2020, some immunoglobulin (IG) lots from US plasma contained neutralizing antibodies against the newly emerged SARS-CoV-2. Paralleled by the increasing numbers of post-COVID-19 donors, IG lot antibody positivity increased to 93% by January 2021, at a mean titer of ~30 IU/mL. The correlation predicted anti-SARS-CoV-2 potency to reach 345 IU/mL by July 2021. In addition to post-COVID-19 donors, the rapidly increasing number of COVID-19 vaccinated plasma donors now result in a mean antibody titer of >600 IU/mL already in July 2021 IG lots, with SARS-CoV-2 antibody titers for several lots even higher than earlier produced hyperimmune globulin products.
Article
Full-text available
Background Several ABO blood groups have been associated with the likelihood of infection, severity, and/or outcome of COVID-19 in hospitalized cohorts, raising the hypothesis that anti-A isoagglutinins in non-A-group recipients could act as neutralizing antibodies against SARS-CoV-2. Materials and methods We run live virus neutralization tests using sera from 58 SARS-CoV-2 seronegative blood donors (27 O-group and 31 A-group) negatives for SARS-CoV-2 IgG to investigate what degree of neutralizing activity could be detected in their sera and eventual correlation with anti-A isoagglutinin titers. Results We could not find clinically relevant neutralizing activity in any blood group, regardless of anti-isoagglutinin titer, Discussion Our findings suggest that mechanisms other than neutralization explain the differences in outcomes from COVID19 seen in different ABO blood groups.
Preprint
The identification of the Omicron variant (B.1.1.529.1 or BA.1) of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) in Botswana in November 20211 immediately raised alarms due to the sheer number of mutations in the spike glycoprotein that could lead to striking antibody evasion. We2 and others3-6 recently reported results in this Journal confirming such a concern. Continuing surveillance of Omicron evolution has since revealed the rise in prevalence of two sublineages, BA.1 with an R346K mutation (BA.1+R346K) and B.1.1.529.2 (BA.2), with the latter containing 8 unique spike mutations while lacking 13 spike mutations found in BA.1. We therefore extended our studies to include antigenic characterization of these new sublineages. Polyclonal sera from patients infected by wild-type SARS-CoV-2 or recipients of current mRNA vaccines showed a substantial loss in neutralizing activity against both BA.1+R346K and BA.2, with drops comparable to that already reported for BA.12,3,5,6. These findings indicate that these three sublineages of Omicron are antigenically equidistant from the wild-type SARS-CoV-2 and thus similarly threaten the efficacies of current vaccines. BA.2 also exhibited marked resistance to 17 of 19 neutralizing monoclonal antibodies tested, including S309 (sotrovimab)7, which had retained appreciable activity against BA.1 and BA.1+R346K2-4,6 . This new finding shows that no presently approved or authorized monoclonal antibody therapy could adequately cover all sublineages of the Omicron variant.