Abstract
Coronavirus disease 2019 (COVID-19) convalescent plasma (CCP) is a safe and effective treatment for COVID-19 in immunocompromised (IC) patients. IC patients have a higher risk of persistent infection, severe disease, and death from COVID-19. Despite the continued clinical use of CCP to treat IC patients, the optimal dose, frequency/schedule, and duration of CCP treatment has yet to be determined, and related best practices guidelines are lacking. A group of individuals with expertise spanning infectious diseases, virology and transfusion medicine was assembled to render an expert opinion statement pertaining to the use of CCP for IC patients. For optimal effect, CCP should be recently and locally collected to match circulating variant. CCP should be considered for the treatment of IC patients with acute and protracted COVID-19; dosage depends on clinical setting (acute vs protracted COVID-19). CCP containing high-titer severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies, retains activity against circulating SARS-CoV-2 variants, which have otherwise rendered monoclonal antibodies ineffective.
Keywords: passive immunization, plasma, antibodies, immunocompromised host
This viewpoint offers expert guidance for the use of coronavirus disease 2019 (COVID-19) convalescent plasma (CCP) to treat immunocompromised patients with acute or protracted COVID-19, addressing indications, dosage, frequency/schedule, and duration of treatment, along with the evidence and associated challenges of use.
Coronavirus disease 2019 (COVID-19) convalescent plasma (CCP) refers to plasma that has been collected from individuals who have recovered from natural severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (with or without SARS-CoV-2 vaccination) [1]. CCP has been shown to be a safe [2] and an effective treatment for COVID-19, when it contains high titers of antibodies against SARS-CoV-2 and is optimally administered early following onset of infection [3–5]. CCP played a significant role in the early public health response to COVID-19. CCP usage diminished following the advent of monoclonal antibodies (mAbs), small molecule antivirals, and—most importantly—preventive strategies (ie, effective vaccines) against COVID-19.
CCP still remains an important therapy for immunocompromised (IC) patients with COVID-19. Although IC patients are a minority (eg, comprising 2.7% of the US population [6]), they are disproportionately affected by COVID-19 [7], with a higher risk of persistent infection and an inability to clear the virus, severe disease and death, despite treatment with small molecule antivirals and/or mAbs [8]. Furthermore, successive waves of viral variants have rendered mAbs ineffective [9]. In contrast, high-titer CCP—by virtue of its polyclonal composition— retains activity against circulating SARS-CoV-2 variants [10]. In acute COVID-19, CCP may be used either in combination with antivirals or alone when alternative options are contraindicated or not readily available. CCP may also be used to treat IC patients with protracted COVID-19 [11–13].
Despite the continued clinical use of CCP to treat IC patients with COVID-19, the optimal timing of its administration, dose, frequency/schedule, and duration of treatment has yet to be determined in IC patients, and related best practices guidelines are lacking.
A General Overview of CCP and Mechanism of Action
Early trials of CCP were disproportionately focused on hospitalized patients with advanced COVID-19, such as those with respiratory distress, hypoxemia, and/or those requiring mechanical ventilation. The findings show that CCP use in “moderate to severe disease does not reduce mortality and has little to no impact on measures of clinical improvement” [14]. By contrast, the early administration of CCP relative to symptom onset or diagnosis of COVID-19 was shown to prevent hospitalization and progression of respiratory disease in the case of outpatient use, as well as improve survival when used early relative to hospital admission [3–5]. Of note, these studies were conducted in largely immunocompetent, unvaccinated subjects with pre-alpha or alpha variant SARS-CoV-2 infection, using plasma that had been collected from donors who had recovered from COVID-19 with pre-Alpha or Alpha variant SARS-CoV-2 [3, 4, 15].
The postulated mechanisms of action of CCP include direct antiviral effects (eg, viral neutralization), viral clearance via immunoglobulin (Ig) M and IgA-mediated neutralization, non-neutralizing IgG Fc-mediated functions (eg, antibody-dependent cellular cytotoxicity, phagocytosis and complement activation) [16], and immunomodulation [17]. CCP is polyclonal, containing thousands of Spike protein-specific antibodies that not only interfere at the receptor binding domain (RBD) but also mediate virus neutralization in non-RBD regions of the Spike protein. This imparts greater durability of CCP to withstand viral evolution [10]. By contrast, mAbs are highly targeted, rendering them vulnerable to mutation: multiple mutations in variants of concern (VOC) have rendered most mAbs ineffective [18]. This risk of viral escape extends to any future mAbs unless the target(s) cannot be mutated or shielded.
Evidence of CCP Use in Immunocompromised Patients With COVID-19
IC patients are at higher risk for poor outcomes after acute COVID-19 as well as protracted COVID-19 symptoms and/or viral persistence (eg, “smoldering” COVID-19), sometimes lasting for months [8, 19].
A systematic review of CCP use in patients with innate or acquired immunosuppression [9] included three randomized clinical trials (collectively representing 214 participants), 5 matched cohort studies (n = 1560 participants) and 138 case reports or case series (n = 623 individuals). These studies were analyzed using a standard fixed effects model that compared the observed deaths among patients transfused with CCP with the expected deaths if all patients were equally at risk [9]. This meta-analysis showed an association between CCP use and a mortality benefit in hospitalized IC patients with COVID-19, and there was a high level of concordance between individual studies. Limitations of this study include its lack of individual patient data, restriction to a single outcome (ie, mortality), differences in the volume and titer of CCP used, and how IC was defined. Nonetheless, the findings from this analysis suggests that CCP is safe and may be beneficial in IC patients with COVID-19.
Numerous case series and case reports also suggest that the effect of CCP in IC patients with COVID-19 is rapid, often achieving viral clearance, improved clinical outcomes and survival [9, 20, 21]. Nonetheless, limitations of these studies (eg, the absence of randomization, a lack of controls, and susceptibility to bias) preclude their use in making a definitive recommendation for CCP.
Evaluation of CCP treatment in IC patients with protracted COVID-19 is limited to 2 case series and 1 case report [11–13]. Clinical improvement was observed in 43 of 49 patients who received CCP. All were treated with other COVID-19-specific therapies; therefore, evaluating the effect of CCP alone is impossible. Of note, 5 of the 49 patients required invasive mechanical ventilation, 4 of whom died despite transfusion of CCP. This is consistent with collective experience that there is limited—if any—clinical benefit of CCP in patients who require invasive mechanical ventilation [14].
Guidelines From Professional Societies and Regulatory Bodies
The US Food and Drug Administration (FDA) authorizes the use of CCP “for the treatment of COVID-19 in patients with immunosuppressive disease or receiving immunosuppressive treatment in either the outpatient or inpatient setting” [22]. Additionally, multiple professional societies provide guidance on CCP in IC patients. AABB (Association of Advancement of Blood and Biotherapies) “suggests CCP transfusion in addition to the usual standard of care for hospitalized patients with COVID-19 and preexisting immunosuppression” yet grades the recommendation as weak with low-certainty evidence [23]. The European conference of infections in leukemia (ECIL-9) and the National Comprehensive Cancer Network (NCCN) also recommend CCP for selected patients, notably for SARS-CoV-2 seronegative hematological patients [24]. The Infectious Diseases Society of America (IDSA) also includes high-titer CCP as a treatment option for outpatients when other options are not possible [25]. The National Institutes of Health (NIH) neither recommends for or against CCP in IC patients [26].
Definition of Immunocompromise
Immunocompromise is a broad term that encompasses a variety of clinical states in which immunity is impaired, including non-modifiable conditions, (eg, advanced age, genetic immunodeficiencies), treatable comorbidities (eg, human immunodeficiency virus [HIV], diabetes, cancer), and iatrogenic states (eg, immunosuppressive and myeloablative therapies). The terms IC, immunosuppression, and immunodeficiency are often used interchangeably. The nature of immunocompromise is often not defined among enrolled participants in CCP trials. Further study is needed to identify which IC patients are most likely to benefit from CCP.
It is biologically plausible that IC individuals who fail to produce an appropriate antibody response to COVID-19 and/or SARS-CoV-2 vaccination are likely to benefit from the SARS-CoV-2 antibodies provided by CCP. Established high risk groups that would qualify for CCP based on this definition include—but are not limited to—organ transplant recipients, patients with auto-immune diseases treated by B-cell depleting agents, and patients with cancer diagnoses, especially those with B-cell deficiency or depletion that may be primary (eg, due to malignancy) or acquired (eg, due to treatment with B-cell depleting agents such as Rituximab) [27, 28]. Practically, these patients are likely to have a minimal or absent response to SARS-CoV-2 vaccination. Testing for the presence of SARS-CoV-2 antibodies could be used to identify suitable candidates for CCP, but there are caveats to this approach: antibody levels may wane with time, be difficult to interpret in patients who have had Evusheld prophylaxis, and may not correlate with CCP functional activity. At present, antibody testing has not been prioritized, impeding its use in clinical decision making. Despite these shortcomings, antibody levels are an objective measure of an individual's ability to mount an immune response and—in the setting of IC—are currently the best biomarker of candidacy for CCP therapy.
DOSAGE, TIMING, AND DURATION DEPEND ON THE INDICATION (Table 1)
Table 1.
Clinical Recommendations
| Target population |
|
| Product qualification |
|
| Initial dose |
|
| Treatment duration |
|
| Frequency |
|
Abbreviations: CCP, convalescent plasma; COVID-19, coronavirus disease 2019; PCR, polymerase chain reaction; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.
Timing of Administration
There are 2 settings when CCP therapy should be considered for IC patients with SARS-CoV-2 infection: (1) acute symptomatic SARS-CoV-2 infection (COVID-19) when there is concern that viral clearance is unlikely (given underlying disease/immune compromise), particularly where there are contraindications to antivirals and limited or no access to effective mAbs; (2) viral persistence in the absence of acute disease (ie, protracted COVID-19), which nevertheless delays return to normal life and/or resumption of full-dose immunosuppression or chemotherapy [29]. Protracted COVID-19 is distinct from “long COVID-19” or post-COVID-19 condition.
CCP Alone or in Combination With Other Therapies
Currently, there is a paucity of data on when or how to administer CCP in concert with other therapies (ie, antiviral therapy) or to comment upon the utility of combination therapy versus CCP alone. Although optimal regimens have yet to be determined, a combination of a direct-acting antiviral plus passive antibody-based therapy (eg, CCP) may be considered for patients with persistent SARS-CoV-2 infection. These patients may not be eligible for nirmatrelvir/ritonavir and molnupiravir (ie, based on duration of symptoms) according to the criteria for emergency use authorization (EUA) in the United States. Therefore, 1 approach for consideration for inpatients with persistent SARS-CoV2 positivity and symptoms is remdesivir plus CCP (eg, 1 unit of CCP every other day × 3 units total if available), again acknowledging that the optimal regimen is unknown.
Dosage and Duration of Treatment
One to 2 units of CCP should be used to treat acute COVID-19 with continued dosing based on clinical response thereafter. In the case of persistent SARS-CoV-2 infection with very high viral loads, multiple doses should be considered at defined intervals until viral clearance is achieved. The group's recommendation acknowledges that the ideal dosage and duration of CCP treatment remains uncertain. The timing and dosage (single vs multiple units) of CCP, should depend on the clinical situation (ie, acute symptomatic vs persistent [infection with few or no symptoms]) and response to previous treatment (eg, failure to clear the virus following treatment with mAbs and/or small molecule antivirals).
Most studies—independent of immune status—pertain to acute COVID-19 in which a dose of 1–2 units of CCP (200–300 mL per unit) has been used; that dose was based on experience with convalescent plasma prior to the COVID-19 pandemic [1]. Two units are more likely to result in detectable antibodies in the recipient than 1 unit alone [30].
Specific to the IC patient population, there is no consensus regarding optimal dosing either in the number of units or dosing frequency. In a systematic review of reported use of CCP in IC patients, the median dose was 2 units (range 1–11 units) [9]. Several cases reports have used multiple units and repeated dosing in IC patients, but there are limited data to support this practice. In the case of dosing multiple units, the risk of transfusion-associated circulatory overload should be considered if administered over short periods, given that IC patients may have concomitant comorbid disease (eg, cardiorespiratory disease and/or renal impairment); however, most patients are likely to tolerate dosages higher than 200 mL [31]. In the case of viral persistence, there is more variability in practice whereby multiple and repeat doses may be administered at different intervals, in an attempt to achieve viral clearance [29, 32].
Determination of Efficacy and Monitoring Outcomes
Monitoring of outcomes should include clinical, laboratory and imaging data; decisions to re-dose should involve an infectious diseases specialist and review of all available data.
Clinical measures include the work of breathing, respiratory rate and oxygen saturation. The radiological response is ascertained by evidence of radiological improvement such as resolution of infiltrates. Although formal laboratory assays of SARS-CoV-2 viral load are preferable when available, sequential cycle threshold (Ct) values can reveal whether CCP treatment had an effect on viral load and in so doing guide recommendations for additional CCP and/or other therapies. Although Ct values are a semi-quantitative measure of viral load and CCP effect, not all laboratories routinely report Ct values, and Ct values are not a standard (ie, approved) clinical test. Some institutions utilize Ct values in decision making; however, different platforms and lack of consistent correlation with viral loads impair interpretation and therefore professional societies have recommended against their use until more data are available. When Ct values are used, the same laboratory/platform is advisable to facilitate interpretation of the results, given the variability between polymerase chain reaction (PCR) platforms. At present, the optimal timing and frequency of Ct determination following CCP transfusion is not known, but in generally 5–7 days seems a reasonable approach for stable patients.
Research and Future Directions
Research is needed to address the knowledge gaps in each of the cited elements governing CCP use, for example, dose, frequency, duration, and monitoring, as well as the interplay between CCP, viral evolution, and the host immune responses. As a first step, we recommend that a registry of immunocompromised CCP recipients be established to obtain high quality observational data.
The US FDA has defined acceptable CCP antibody thresholds for qualification of CCP for clinical use, based on various Spike protein antibody assays [22]. At present, the FDA authorization requires a history of SARS-CoV-2 infection, with a maximum time allowed since that infection, to be eligible to donate CCP. Individuals are not eligible to donate CCP based on a history of SARS-CoV-2 vaccination alone. We recommend that the current algorithm for donor selection be revised given ample evidence that most blood donors are both convalescent and vaccinated [33]. Routine donor-based laboratory screening for SARS-CoV-2 Spike antibodies would simplify workflow considerably.
The CCP collected from individuals who have been vaccinated and recovered from natural COVID-19 [so called “VaxPlasma” or “hybrid plasma”] has 10-times greater titers of antibodies (potentially delivering more antibody neutralization with similar or less volume) against SARS-CoV-2 than standard high-titer CCP [30, 34, 35], upon which initial authorizations of CCP were granted. Moreover, “VaxPlasma” is highly effective against Omicron variants, unlike most mAbs, which have been shown to be ineffective [35–37]. Nonetheless, “VaxPlasma” or hybrid plasma is not yet an approved product and qualification requirements still need to be defined.
A fundamental challenge is the inherent variability in the composition of CCP, whereby it remains uncertain as to whether any two units are of similar dose and efficacy, even when both are collected from the same donor. This may be a contributing factor for negative findings in studies of CCP [38]. Approaches that merit investigation include pooling multiple ABO-identical units to create a more uniform (ie, refined) version of CCP, or transfusing units from different donors.
Logistical Considerations and Challenges
CCP is not being actively collected by most blood services, thus forcing reliance on old stocks of CCP, introducing several challenges. First, CCP that is in use is not temporally or geographically matched to circulating variants and, therefore, may have diminished efficacy [39]. Second, dosing is subject to the availability of CCP and any recommendation to transfuse high doses of CCP to IC patients requires the confidence that multiple units will be available. Similarly, the waning supply with a notable low representation of Group B and AB plasma could restrict CCP to A and O recipients; even if multiple doses were to be prescribed, there may not be sufficient units available. Despite a lack of consensus on whether out-of-group plasma should be allowed, examples of how transfusion with ABO incompatible plasma could be undertaken safely, have been described [40].
CCP is optimally effective when transfused early relative to onset of symptoms or diagnosis [3, 4]. This highlights the need for rapid diagnostics, timely referral for treatment, and—ideally—capacity for outpatient transfusion (CCP is mainly being administered in an inpatient setting). There are challenges of outpatient or home administration CCP [41]. However, with at-home testing, mAbs have been administered in patients’ homes to prevent hospitalization. Outpatient facilities that have been used to infuse mAbs could be repurposed for CCP transfusions, in collaboration with the local transfusion services [41]. Of note, IC patients are still at risk of disease progression despite early intervention, whereby later administration of CCP may still be beneficial, contrary to the recommendations for those who are immunocompetent.
There are inherent challenges that are especially pertinent to IC patients. These include the inability to conduct traditional randomized controlled trials in this patient population. Although formal evidence-based clinical guidelines are needed to address the oversight and approval of CCP, it is impractical to conduct new trials of CCP each time a novel variant or new CCP collection workflow arises. Instead, it is reasonable to apply in vitro data, as has occurred with the mAbs.
CONCLUSION
In summary, there is a growing body of evidence for the role of CCP in the treatment of IC patients with COVID-19. Our view is consistent with and supported by recommendations of other learned groups such as AABB, NCCN, and ECIL-9 [23, 24]. Despite knowledge gaps, our panel recommends that CCP be considered for treatment of IC patients as follows: dosing (number of units and frequency) should be tailored to the indication, for example, acute symptomatic COVID-19, protracted COVID-19 (persistent SARS-CoV-2 positivity). For IC patients with acute COVID, CCP should be considered but the role of combination therapy (ie, with antivirals) is not yet known. For patients with persistent infection, transfusion of CCP should occur at regularly scheduled intervals along with antivirals. Clinical outcomes, for example, improvement in symptoms for patients with acute and/or protracted disease, should be used to guide treatment along with Ct values when available. Finally, there are regulatory and logistical barriers to the use of CCP. Given that data show that the majority of donors are now vaccinated and/or have a history of natural SARS-CoV-2 infection, there is a case to revisit the regulatory requirements for donation and qualification of CCP, which would simplify procurement, thus improving its availability.
Notes
Financial support. E. M. B.'s effort is supported in part by the National Heart Lung and Blood Institute (grant number 1K23HL151826). K. G. reports support for this work made to institution from Octapharma, US Department of Defense's Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense, National Institutes of Health (NIH) National Institute of Allergy and Infectious Diseases (grant number 3R01AI152078-01S1), NIH National Center for Advancing Translational Sciences (grant numbers U24TR001609-S3 and UL1TR00309), Mental Wellness Foundation, HealthNetwork Foundation, Bloomberg Philanthropies, Moriah Fund, Shear family foundation, State of Maryland, Defense Health Agency (grant number W911QY2090012). J. P. H. reports unrestricted gift to Washington University to administer to Henderson laboratory to support activities related to COVID-19 convalescent plasma from Octapharma of Paramus, New Jersey, USA.
Contributor Information
Evan M Bloch, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Daniele Focosi, North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy.
Shmuel Shoham, Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Jonathon Senefeld, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Aaron A R Tobian, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Lindsey R Baden, Department of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA.
Pierre Tiberghien, Etablissement Français du Sang, La Plaine-St-Denis and Université de Franche-Comté, Besançon, France.
David J Sullivan, Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
Claudia Cohn, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA.
Veronica Dioverti, Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Jeffrey P Henderson, Departments of Internal Medicine (Division of Infectious Diseases) and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
Cynthia So-Osman, Department Transfusion Medicine, Division Blood Bank, Sanquin Blood Supply Foundation, Amsterdam, The Netherlands; Department Haematology, Erasmus Medical Centre, Rotterdam, The Netherlands.
Justin E Juskewitch, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester campus, Minnesota, USA.
Raymund R Razonable, Department of Internal Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA.
Massimo Franchini, Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy.
Ruchika Goel, Division of Hematology/Oncology, Simmons Cancer Institute at SIU School of Medicine and Mississippi Valley Regional Blood Center, Springfield, Illinois, USA.
Brenda J Grossman, Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
Arturo Casadevall, Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
Michael J Joyner, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA.
Robin K Avery, Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
Liise-anne Pirofski, Department of Medicine, Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York, USA.
Kelly A Gebo, Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
References
- 1. Bloch EM, Shoham S, Casadevall A, et al. Deployment of convalescent plasma for the prevention and treatment of COVID-19. J Clin Invest 2020; 130:2757–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Joyner MJ, Bruno KA, Klassen SA, et al. Safety update: cOVID-19 convalescent plasma in 20,000 hospitalized patients. Mayo Clin Proc 2020; 95:1888–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Libster R, Pérez Marc G, Wappner D, et al. Early high-titer plasma therapy to prevent severe COVID-19 in older adults. N Engl J Med 2021; 384:610–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Sullivan DJ, Gebo KA, Shoham S, et al. Early outpatient treatment for COVID-19 with convalescent plasma. N Engl J Med 2022; 386:1700–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Joyner MJ, Carter RE, Senefeld JW, et al. Convalescent plasma antibody levels and the risk of death from COVID-19. N Engl J Med 2021; 384:1015–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Harpaz R, Dahl RM, Dooling KL. Prevalence of immunosuppression among US adults, 2013. JAMA 2016; 316:2547–8. [DOI] [PubMed] [Google Scholar]
- 7. Booth A, Reed AB, Ponzo S, et al. Population risk factors for severe disease and mortality in COVID-19: a global systematic review and meta-analysis. PLoS One 2021; 16:e0247461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Dioverti V, Salto-Alejandre S, Haidar G. Immunocompromised patients with protracted COVID-19: a review of “long persisters.” Curr Transpl Rep 2022; 9:209–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Senefeld JW, Franchini M, Mengoli C, et al. COVID-19 Convalescent plasma for the treatment of immunocompromised patients: a systematic review and meta-analysis. JAMA Netw Open 2023; 6:e2250647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Sullivan DJ, Franchini M, Joyner MJ, Casadevall A, Focosi D. Analysis of anti-SARS-CoV-2 Omicron-neutralizing antibody titers in different vaccinated and unvaccinated convalescent plasma sources. Nat Commun 2022; 13:6478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hueso T, Pouderoux C, Péré H, et al. Convalescent plasma therapy for B-cell-depleted patients with protracted COVID-19. Blood 2020; 136:2290–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Ripoll JG, Gorman EK, Juskewitch JE, et al. Vaccine-boosted convalescent plasma therapy for patients with immunosuppression and COVID-19. Blood Adv 2022; 6:5951–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Ordaya EE, Abu Saleh OM, Stubbs JR, Joyner MJ. Vax-plasma in patients with refractory COVID-19. Mayo Clin Proc 2022; 97:186–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Piechotta V, Iannizzi C, Chai KL, et al. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: a living systematic review. Cochrane Database Syst Rev 2021; 5:Cd013600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Gharbharan A, Jordans C, Zwaginga L, et al. Outpatient convalescent plasma therapy for high-risk patients with early COVID-19: a randomized placebo-controlled trial. Clin Microbiol Infect 2023; 29:208–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Natarajan H, Crowley AR, Butler SE, et al. Markers of polyfunctional SARS-CoV-2 antibodies in convalescent plasma. mBio 2021; 12:e00765-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Rojas M, Rodríguez Y, Monsalve DM, et al. Convalescent plasma in COVID-19: possible mechanisms of action. Autoimmun Rev 2020; 19:102554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Takashita E, Yamayoshi S, Fukushi S, et al. Efficacy of antiviral agents against the Omicron subvariant BA.2.75. N Engl J Med 2022; 387:1236–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Alasfar S, Chiang TP, Snyder AJ, et al. PASC in solid organ transplant recipients with self-reported SARS-CoV-2 infection. Transplantation 2022; 107:181–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Senefeld JW, Klassen SA, Ford SK, et al. Use of convalescent plasma in COVID-19 patients with immunosuppression. Transfusion 2021; 61:2503–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Hueso T, Godron AS, Lanoy E, et al. Convalescent plasma improves overall survival in patients with B-cell lymphoid malignancy and COVID-19: a longitudinal cohort and propensity score analysis. Leukemia 2022; 36:1025–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. FDA . Investigational COVID-19 Convalescent Plasma Guidance for Industry. Accessed January 8.
- 23. Estcourt LJ, Cohn CS, Pagano MB, et al. Clinical practice guidelines from the Association for the Advancement of Blood and Biotherapies (AABB): COVID-19 convalescent plasma. Ann Intern Med 2022; 175:1310–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Cesaro S, Ljungman P, Mikulska M, et al. Recommendations for the management of COVID-19 in patients with haematological malignancies or haematopoietic cell transplantation, from the 2021 European Conference on Infections in Leukaemia (ECIL 9). Leukemia 2022; 36:1467–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. IDSA . IDSA Guideline on the Treatment and Management of COVID-19. Available at:https://www.idsociety.org/covid-19-real-time-learning-network/therapeutics-and-interventions/convalescent-plasma/#. Accessed August 14.
- 26. NIH . COVID-19 Treatment Guidelines. Available at:https://www.covid19treatmentguidelines.nih.gov/management/clinical-management-of-adults/. Accessed November 4.
- 27. Denkinger CM, Janssen M, Schäkel U, et al. Anti-SARS-CoV-2 antibody containing plasma improves outcome in patients with hematologic or solid cancer and severe COVID-19 via increased neutralizing antibody activity: a randomized clinical trial. Nat Cancer 2023; 4:96–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Gachoud D, Pillonel T, Tsilimidos G, et al. Antibody response and intra-host viral evolution after plasma therapy in COVID-19 patients pre-exposed or not to B-cell-depleting agents. Br J Haematol 2022; 199:549–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Focosi D, Senefeld JW, Joyner MJ, et al. Lower anti-spike levels in B-cell-depleted patients after convalescent plasma transfusion suggest the need for repeated doses. Br J Haematol 2023; 200:e22–4. [DOI] [PubMed] [Google Scholar]
- 30. Leon J, Merrill AE, Rogers K, et al. SARS-CoV-2 antibody changes in patients receiving COVID-19 convalescent plasma from normal and vaccinated donors. Transfus Apher Sci 2022; 61:103326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Senefeld JW, Johnson PW, Kunze KL, et al. Access to and safety of COVID-19 convalescent plasma in the United States expanded access program: a national registry study. PLoS Med 2021; 18:e1003872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Belcari G, Conti A, Mazzoni A, Lanza M, Mazzetti P, Focosi D. Clinical and virological response to convalescent plasma in a chronic lymphocytic leukemia patient with COVID-19 pneumonia. Life (Basel) 2022; 12:1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Jones JM, Opsomer JD, Stone M, et al. Updated US infection- and vaccine-induced SARS-CoV-2 seroprevalence estimates based on blood donations, July 2020–December 2021. JAMA 2022; 328:298–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Vickers MA, Sariol A, Leon J, et al. Exponential increase in neutralizing and spike specific antibodies following vaccination of COVID-19 convalescent plasma donors. Transfusion 2021; 61:2099–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Li M, Beck EJ, Laeyendecker O, et al. Convalescent plasma with a high level of virus-specific antibody effectively neutralizes SARS-CoV-2 variants of concern. Blood Adv 2022; 6:3678–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Sullivan D, Franchini M, Joyner M, Casadevall A, Focosi D. Analysis of anti-SARS-CoV2 Omicron neutralizing antibody titers in different vaccinated and unvaccinated convalescent plasma sources. Nat Commun 2022; 13:6478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Sullivan DJ, Franchini M, Senefeld JW, Joyner MJ, Casadevall A, Focosi D. Plasma after both SARS-CoV-2 boosted vaccination and COVID-19 potently neutralizes BQ.1.1 and XBB.1. bioRxiv [Preprint]. December 16, 2022. Available at: 10.1101/2022.11.25.517977. [DOI] [PMC free article] [PubMed]
- 38. Korley FK, Durkalski-Mauldin V, Yeatts SD, et al. Early convalescent plasma for high-risk outpatients with COVID-19. N Engl J Med 2021; 385:1951–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Kunze KL, Johnson PW, van Helmond N, et al. Mortality in individuals treated with COVID-19 convalescent plasma varies with the geographic provenance of donors. Nat Commun 2021; 12:4864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Focosi D, Farrugia A. ABO-incompatible convalescent plasma transfusion: yes, you can. Transfusion Med 2021; 31:215–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Bloch EM, Tobian AAR, Shoham S, et al. How do I implement an outpatient program for the administration of convalescent plasma for COVID-19? Transfusion 2022; 62:933–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
