9
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: not found
      • Article: not found

      Understanding the parameters guiding the best practice for treating B‐cell‐depleted patients with COVID‐19 convalescent plasma therapy

      letter

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The commentary by Focosi et al. 1 highlights two key aspects of the patient‐blood management for coronavirus disease 2019 (COVID‐19) convalescent plasma (CCP) therapy in seronegative patients who are pre‐exposed to B‐cell‐depleting agents. The first one relates to the qualitative composition of CCP used for patient treatment. The efficacy of CCP is strongly affected by the titres of neutralising antibodies (nAb) present at the time of donation. 2 , 3 Moreover, there is growing evidence that the nAb activity of plasma units collected from unvaccinated donors infected during the former COVID‐19 waves is reduced against novel variants such as Omicron. 4 Hence, the efficacy of collected plasma is likely evolving alongside the exposure of potential donors to successive variant outbreaks and vaccination campaigns against severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2). At the beginning of the pandemic, CCP was used as a front‐line treatment; however, its wide heterogeneity in nAb titres made it difficult to recruit suitable donors of high neutralising titres, as defined by the United States Food and drug Administration (FDA). In contrast, nAb induced after two doses of mRNA vaccination have generally higher and more homogenous titres, which further remain effective against Omicron following a booster dose. 5 From March to July 2021, we treated 19 immunosuppressed patients with plasma collected from COVID‐naive two‐dose mRNA‐vaccinated donors, under a compassionate use protocol, according to the Swiss regulation rules. 6 No relevant difference was observed between patients receiving CCP (n = 17) versus vaccinated plasma (VP, n = 19), indicating that VP therapy is safe and effective in patients with B‐cell lymphopenia. In the meantime, CCP donors are increasingly getting vaccinated, and three‐dose VP donors are being boosted by a breakthrough infection (defined as ‘hybrid plasma’, Vax‐CCP or CP/VP). Their plasmas contain higher nAb titres and broader antibody specificity than CCP, 4 , 7 , 8 as shown by the increase in anti‐spike immunoglobulin G titres between 2021 and 2022 (Figure 1A). Thus, Vax‐CCP is emerging as the most convenient available source of polyclonal antibody plasma, given the failure of many monoclonal antibody therapies, due to the Omicron immune escape variants. 9 FIGURE 1 SARS‐CoV‐2 viral load response follow‐up in B‐cell‐depleted patients (n = 11) treated with plasma therapy (from December 2021 through September 2022). All patients had received anti‐CD20 therapy and, except of one patient, were hospitalised for moderate to severe COVID‐19. A favourable clinical outcome was observed in 10 out the 11 plasma‐treated patients. (A) Anti‐spike (anti‐S) protein IgG titres were collected from COVID‐19‐experienced only plasma donors (2020, October 2020–February 2021) or COVID‐19‐experienced vaccine‐boosted (VaxCCP or CP/VP) plasma donors (2021, June–August 2021; 2022, February–September 2022). (B) Comparison of anti‐S IgG antibody titres in each patient before and after 1 × CP/VP or 2 × CP/VP treatment, using an in‐house developed Luminex assay. 11 The maximum reached value is depicted. (C) The PANGO lineage is indicated for each patient. (D) SARS‐CoV‐2 RNA detection 12 in nasopharyngeal (NP) swabs (copies/ml) before (Day –5 to Day 0) treatment and over‐time kinetics after treatment with 1 × CP/VP (1×) or 2 × CP/VP (2×). The p value is by Mann–Whitney test. (E) Time‐to‐negativity of the NP swabs in patients treated with 1 × CP/VP (1×) or 2 × CP/VP (2×). CCP, COVID‐19 convalescent plasma; COVID‐19, coronavirus disease 2019; IgG, immunoglobulin G; n.s., not significant; PANGO, Phylogenetic Assignment of Named Global Outbreak; SARS‐CoV‐2, severe acute respiratory syndrome coronavirus‐2; VP, vaccinated plasma. The second key aspect commented upon by Focosi et al 1 concerns the best practice for treating immunocompromised seronegative patients with COVID‐19 with plasma therapy. It still remains to be defined which therapeutic plasma protocol is optimal to achieve a rapid and complete viral response. 10 In this regard, Focosi et al 1 propose to start with a loading Vax‐CCP dose of 600 ml followed by weekly 600‐ml dose transfusion, allowing for a faster viral clearance in patients than those receiving a single‐dose unit. In our centre, we use an initial 400‐ml dose of Vax‐CCP issued from two vaccinated and recently‐infected donors having recovered from a contemporary viral strain. In case of insufficient clinical and/or viral load response, we treat again with the same plasma dose but from other convalescent vaccine‐boosted individuals. Using this strategy, we recently followed the SARS‐CoV‐2 viral load response in 11 B‐cell‐depleted antibody‐negative patients with a documented onco‐haematological (seven patients) or autoimmune disorder (four patients), after plasma therapy. Patients were classified according to whether they had Vax‐CCP treatment once or twice (Figure 1B) and presented various Phylogenetic Assignment of Named Global Outbreak (PANGO) lineages, depending on the predominant circulating strain(s) at the time of diagnosis (Figure 1C). Our data indicate that seven out nine patients were able to clear the virus within the same time‐range (<35 days, Figure 1D,E) as reported for patients with endogenous anti‐SARS‐CoV‐2 responses, 6 among them two had received two serial transfusions. Hence, our tailored approach is consistent with the relative low number of refractory cases (six of 36) previously observed 6 and enables sparing Vax‐CCP resources of high anti‐SARS‐CoV‐2 titres, which heavily rely on the continuous collection of donor plasma. Understanding which parameter(s) may be predictive of complete viral clearance would likely help adjusting Vax‐CCP treatment. As such, the question of whether basal viral load is typically higher in B‐cell‐depleted patients than in B‐cell‐undepleted ones and thus represents a red‐flag could alas not be addressed in the dataset initially reported, 6 as both cohorts differed substantially in terms of the underlying viral variant (pre‐alpha vs. alpha predominance respectively). Still, we recently observed that patients transfused twice with Vax‐CCP had higher levels of initial SARS‐CoV‐2 viral loads than those receiving only one‐time plasma (Figure 1D). These observations suggest that viral loads before Vax‐CCP treatment may potentially define those seronegative patients more at risk of delayed clinical recovery and/or viral clearance, and who would require additional transfusions. Importantly, randomised controlled trials, like the recently reopened Randomised, Embedded, Multifactorial, Adaptive Platform Trial for Community‐Acquired Pneumonia (REMAP‐CAP) trial, specifically focusing on immunocompromised patients treated with Vax‐CCP, should further help addressing such important points, paving future guidelines for the use of therapeutic plasma. AUTHOR CONTRIBUTIONS Nathalie Rufer wrote the first draft; David Gachoud, Claire Bertelli and Nathalie Rufer revised the manuscript. CONFLICT OF INTEREST The authors declare that they have no competing interests. PATIENT CONSENT STATEMENT Each patient provided informed consent for plasma transfusion and data collection.

          Related collections

          Most cited references13

          • Record: found
          • Abstract: found
          • Article: not found

          Early High-Titer Plasma Therapy to Prevent Severe Covid-19 in Older Adults

          Abstract Background Therapies to interrupt the progression of early coronavirus disease 2019 (Covid-19) remain elusive. Among them, convalescent plasma administered to hospitalized patients has been unsuccessful, perhaps because antibodies should be administered earlier in the course of illness. Methods We conducted a randomized, double-blind, placebo-controlled trial of convalescent plasma with high IgG titers against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in older adult patients within 72 hours after the onset of mild Covid-19 symptoms. The primary end point was severe respiratory disease, defined as a respiratory rate of 30 breaths per minute or more, an oxygen saturation of less than 93% while the patient was breathing ambient air, or both. The trial was stopped early at 76% of its projected sample size because cases of Covid-19 in the trial region decreased considerably and steady enrollment of trial patients became virtually impossible. Results A total of 160 patients underwent randomization. In the intention-to-treat population, severe respiratory disease developed in 13 of 80 patients (16%) who received convalescent plasma and 25 of 80 patients (31%) who received placebo (relative risk, 0.52; 95% confidence interval [CI], 0.29 to 0.94; P=0.03), with a relative risk reduction of 48%. A modified intention-to-treat analysis that excluded 6 patients who had a primary end-point event before infusion of convalescent plasma or placebo showed a larger effect size (relative risk, 0.40; 95% CI, 0.20 to 0.81). No solicited adverse events were observed. Conclusions Early administration of high-titer convalescent plasma against SARS-CoV-2 to mildly ill infected older adults reduced the progression of Covid-19. (Funded by the Bill and Melinda Gates Foundation and the Fundación INFANT Pandemic Fund; Dirección de Sangre y Medicina Transfusional del Ministerio de Salud number, PAEPCC19, Plataforma de Registro Informatizado de Investigaciones en Salud number, 1421, and ClinicalTrials.gov number, NCT04479163.)
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection

            Emerging SARS-CoV-2 variants have raised concerns about resistance to neutralizing antibodies elicited by previous infection or vaccination. We examined whether sera from recovered and nave donors collected prior to, and following immunizations with existing mRNA vaccines, could neutralize the Wuhan-Hu-1 and B.1.351 variants. Pre-vaccination sera from recovered donors neutralized Wuhan-Hu-1 and sporadically neutralized B.1.351, but a single immunization boosted neutralizing titers against all variants and SARS-CoV-1 by up to 1000-fold. Neutralization was due to antibodies targeting the receptor binding domain and was not boosted by a second immunization. Immunization of nave donors also elicited cross-neutralizing responses, but at lower titers. Our study highlights the importance of vaccinating both uninfected and previously infected persons to elicit cross-variant neutralizing antibodies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Convalescent Plasma Antibody Levels and the Risk of Death from Covid-19

              Abstract Background Convalescent plasma has been widely used to treat coronavirus disease 2019 (Covid-19) under the presumption that such plasma contains potentially therapeutic antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that can be passively transferred to the plasma recipient. Whether convalescent plasma with high antibody levels rather than low antibody levels is associated with a lower risk of death is unknown. Methods In a retrospective study based on a U.S. national registry, we determined the anti–SARS-CoV-2 IgG antibody levels in convalescent plasma used to treat hospitalized adults with Covid-19. The primary outcome was death within 30 days after plasma transfusion. Patients who were enrolled through July 4, 2020, and for whom data on anti–SARS-CoV-2 antibody levels in plasma transfusions and on 30-day mortality were available were included in the analysis. Results Of the 3082 patients included in this analysis, death within 30 days after plasma transfusion occurred in 115 of 515 patients (22.3%) in the high-titer group, 549 of 2006 patients (27.4%) in the medium-titer group, and 166 of 561 patients (29.6%) in the low-titer group. The association of anti–SARS-CoV-2 antibody levels with the risk of death from Covid-19 was moderated by mechanical ventilation status. A lower risk of death within 30 days in the high-titer group than in the low-titer group was observed among patients who had not received mechanical ventilation before transfusion (relative risk, 0.66; 95% confidence interval [CI], 0.48 to 0.91), and no effect on the risk of death was observed among patients who had received mechanical ventilation (relative risk, 1.02; 95% CI, 0.78 to 1.32). Conclusions Among patients hospitalized with Covid-19 who were not receiving mechanical ventilation, transfusion of plasma with higher anti–SARS-CoV-2 IgG antibody levels was associated with a lower risk of death than transfusion of plasma with lower antibody levels. (Funded by the Department of Health and Human Services and others; ClinicalTrials.gov number, NCT04338360.)
                Bookmark

                Author and article information

                Contributors
                nathalie.rufer@unil.ch
                Journal
                Br J Haematol
                Br J Haematol
                10.1111/(ISSN)1365-2141
                BJH
                British Journal of Haematology
                John Wiley and Sons Inc. (Hoboken )
                0007-1048
                1365-2141
                10 November 2022
                January 2023
                10 November 2022
                : 200
                : 2 ( doiID: 10.1111/bjh.v200.2 )
                : e25-e27
                Affiliations
                [ 1 ] Department of Internal Medicine Lausanne University Hospital and University of Lausanne Lausanne Switzerland
                [ 2 ] Medical Education Unit, School of Medicine, Faculty of Biology and Medicine University of Lausanne Lausanne Switzerland
                [ 3 ] Institute of Microbiology Lausanne University Hospital and University of Lausanne Lausanne Switzerland
                [ 4 ] Interregional Blood Transfusion SRC Epalinges Switzerland
                [ 5 ] Department of Oncology Lausanne University Hospital and University of Lausanne Epalinges Switzerland
                Author notes
                [*] [* ] Correspondence

                Nathalie Rufer, Department of Oncology, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland.

                Email: nathalie.rufer@ 123456unil.ch

                Author information
                https://orcid.org/0000-0002-9871-6271
                Article
                BJH18540 BJH-2022-01988.R1
                10.1111/bjh.18540
                9877798
                36354067
                4101bf60-f725-4575-8429-f8660d02eef1
                © 2022 British Society for Haematology and John Wiley & Sons Ltd.

                This article is being made freely available through PubMed Central as part of the COVID-19 public health emergency response. It can be used for unrestricted research re-use and analysis in any form or by any means with acknowledgement of the original source, for the duration of the public health emergency.

                History
                : 19 October 2022
                : 12 October 2022
                : 19 October 2022
                Page count
                Figures: 1, Tables: 0, Pages: 3, Words: 1583
                Categories
                Letter to the Editor
                Letters to the Editor–Online Only
                Custom metadata
                2.0
                January 2023
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.2.4 mode:remove_FC converted:26.01.2023

                Hematology
                anti‐cd20 therapy,covid‐19 convalescent plasma,immunocompromised,sars‐cov‐2 viral loads

                Comments

                Comment on this article