Skip to main content

Convalescent plasma to treat critically ill patients with COVID-19: framing the need for randomised clinical trials

We are in a severe acute respiratory distress syndrome coronavirus 2 (SARS-CoV-2) pandemic, causing coronavirus disease (COVID-19). SARS-CoV-2 is an enveloped RNA virus with cell entry facilitated by spike (S) protein that has a cleavage site at the S1–S2 boundary and other structural proteins such as membrane (M), envelope (E), and nucleocapsid (N) proteins [1]. Currently, there are two lineages of SARS-CoV-2 virus infecting humans, with similar virulence and clinical outcomes, derived from a common ancestor that originated in December 2019 in Wuhan [1, 2]. Most patients who recover from SARS-CoV-2 illness will develop antibodies and memory lymphocytes against these proteins, which gives them immunity [3]. In this editorial, we discuss the biological, operational, and methodological questions that arise when designing a randomised controlled trial (RCT) of convalescent plasma in COVID-19.

What is convalescent plasma therapy?

Convalescent plasma refers to acellular plasma fraction of blood, containing antibodies against SARS-CoV-2 antigens, with virus neutralisation properties, collected from patients who have recovered from SARS-CoV-2 infections. Passive immunisation with ABO blood group-compatible convalescent plasma will reduce viral burden as neutralising antibodies will binding to the viral spike protein to either prevent interaction with angiotensin-converting enzyme-2 receptor or block the conformational changes in spike protein preventing fusion to host cell membrane and provide immunomodulation.

What do we know thus far about convalescent plasma therapy in COVID-19 illness?

Since the recent Cochrane review that highlighted very low-certainty evidence on the effectiveness and safety of convalescent plasma in COVID-19 patients [4], Joyner and colleagues have reported safety results from a compassionate use convalescent plasma therapy programme in 5000 adults with COVID-19. They highlight that convalescent plasma is a safe treatment with an overall serious adverse event rate of < 1% (n = 36 events), with TACO occuring in 7 patients, TRALI in 11 patients, and allergic transfusion reaction in 3 patients [5]. To date, one RCT has been published. This open-label trial stopped early after recruiting 103 of a planned 200 patients sample size were enrolled. The stoppage was due to low patient recruitment, as the pandemic abated in China, and importantly not for safety reasons [6]. The participants had either severe (respiratory distress and/or hypoxemia) or life-threatening (shock, organ failure, or requiring mechanical ventilation) COVID-19 illness. The intervention, ABO-compatible convalescent plasma at a dose of 4 to 13 ml/kg of recipient body weight, and with an S-RBD-specific IgG titre of at least 1:640. The primary outcome was time to clinical improvement within 28 days, defined as patient discharged alive or reduction of 2 points on a 6-point disease severity scale. The overall trial result was no statistically significant improvement in time to clinical improvement within 28 days between convalescent plasma with standard of care versus standard of care alone. However, any inference from this trial is limited by it's early termination.

Why do we need more RCTs of convalescent plasma?

The risks of administering plasma screened for common blood-borne pathogens are small, but include allergy/anaphylaxis, transfusion-related acute lung injury (TRALI), and transfusion-associated circulatory overload (TACO) [7]. TRALI and TACO are relevant as many COVID-19 patients have incipient respiratory failure that may worsen with convalescent plasma transfusion-related volume loading. Another specific concern with this intervention is antibody-dependent enhancement (ADE). In SARS-1 coronaviruses, ADE occurs by S protein neutralising antibodies enhancing viral entry into cells though fragment-crystallisable (Fc) receptor expressing cells such as monocytes [8]. This has been shown to worsen lung injury in SARS-1 patients [9]. Non-randomised clinical use (compassionate) will not provide evidence of efficacy, which is an important consideration, as passive immunotherapy was ineffective in severe influenza A [10], and Ebola [11]. The impact of these harms would be difficult to identify outside a well-conducted RCT that collects adverse event data in a standardised way, whilst answering the efficacy question.

Can we rapidly provide convalescent plasma with neutralising antibodies during a pandemic?

Convalescent plasma can be collected safely from individuals who have recovered from laboratory-confirmed SARS-CoV-2 infection, as neutralising antibody responses begin by 14 days and continue to increase over the next few weeks. Currently, it is uncertain how long these antibodies persist, but in other coronavirus infections, neutralising antibodies may persist at high titres for at least 3 months before declining [12]. Therefore, collection of plasma around 28 days after recovery will provide an effective product with high titres of neutralising antibodies.

However, neither the method to assess viral neutralisation ability of convalescent plasma prior to administration nor the minimum titre of neutralising antibody that is required for treating critically ill patients with COVID-19 is known. There are two methods to assess viral neutralisation ability—pseudotype and live-virus assays. Pseudotype assays using harmless viruses that express the coronavirus spike protein, the target of neutralising antibodies, are a safer, easier, and more sensitive method for detecting neutralising antibody than live-virus assays that assess neutralisation of invasion of tissue culture cells by live virus [13]. The titres of antibody dose vary between studies, from 400 ml of ABO-compatible convalescent plasma with neutralising antibody titre > 1:40 [14] to single 200 ml dose of inactivated convalescent plasma with neutralising antibody titre > 1:640 [15].

What are the key design issues to consider in RCTs of convalescent plasma?

Current trials include participants with a range of COVID-19 illness spectrum, the intervention (convalescent plasma different timing, different doses, and need for molecular evidence of viral infection) and comparators are different, ranging from standard of care to use of regular plasma for blinding that adds transfusion-related risks in comparator population, and outcomes differ between trials. It is conceivable that the treatment effect of convalescent plasma may differ by illness severity, by dose in terms of volume, concentration of neutralisation antibody, and the risk of ADE along with other adverse events during COVID-19 illness (Table 1) [4].

Table 1 Ongoing randomised controlled trials of convalescent plasma in COVID-19 illness assessed using the PICO framework. These RCTs were identified in a recent Cochrane review by Valk et al. [4]. Participants: We report the setting (severely ill/critically ill versus general wards). In high-risk non-ventilated patients (high inspired oxygen, and/or non-invasive ventilation), this could reduce the need for mechanical ventilation. In ventilated patients, this may translate into improved mortality and reduced length of critical care stay. Intervention: For intervention, we report the description of convalescent plasma volume and titres if highlighted. In SARS-1 patients, convalescent plasma improved outcomes when administered within 14 days of illness onset and in those without detectable antibodies against coronavirus at the time of infusion. Only four studies use a predetermined neutralising titre cutoff with convalescent plasma. Comparator: We highlight whether the ordinary plasma or standard of care was chosen. In five RCTs, the comparator is ordinary plasma transfusion, which may enhance blinding but comes with risks of blood product. When summarising the ongoing current trials, it is unlikely that an efficacy signal would be generated from many of these trials due their methodological limitations (such as small sample size) and biological limitations (such as lack of pre-defined cutoff for neutralising antibody titres). For outcome, we list only the primary outcome for the trial. We also highlight the proposed sample size in the trial.

In summary, there is a clear biological framework for considering convalescent plasma as a potential intervention in COVID-19 illness. However, we need high-quality randomised controlled trials prior to using convalescent plasma as standard care in SARS-CoV-2 infections.

Availability of data and materials

Not applicable.

References

  1. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. The proximal origin of SARS-CoV-2. Nat Med. 2020;26(4):450–2.

    Article  CAS  Google Scholar 

  2. Zhang X, Tan Y, Ling Y, Lu G, Liu F, Yi Z, Jia X, Wu M, Shi B, Xu S, et al. Viral and host factors related to the clinical outcome of COVID-19. Nature. 2020. https://www.nature.com/articles/s41586-020-2355-0.

  3. Ni L, Ye F, Cheng ML, Feng Y, Deng YQ, Zhao H, Wei P, Ge J, Gou M, Li X, et al. Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity. 2020;52(6):971–7 e973.

    Article  CAS  Google Scholar 

  4. Valk SJ, Piechotta V, Chai KL, Doree C, Monsef I, Wood EM, Lamikanra A, Kimber C, McQuilten Z, So-Osman C, et al. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: a rapid review. Cochrane Database Syst Rev. 2020;5:CD013600.

    PubMed  Google Scholar 

  5. Joyner MJ, Wright RS, Fairweather D, Senefeld JW, Bruno KA, Klassen SA, Carter RE, Klompas AM, Wiggins CC, Shepherd JR, et al. Early safety indicators of COVID-19 convalescent plasma in 5,000 patients. J Clin Invest. 2020. https://www.nature.com/articles/s41586-020-2355-0.

  6. Li L, Zhang W, Hu Y, Tong X, Zheng S, Yang J, Kong Y, Ren L, Wei Q, Mei H, et al. Effect of convalescent plasma therapy on time to clinical improvement in patients with severe and life-threatening COVID-19: a randomized clinical trial. JAMA. 2020. https://www.nature.com/articles/s41586-020-2355-0.

  7. MacLennan S, Williamson LM. Risks of fresh frozen plasma and platelets. J Trauma. 2006;60(6 Suppl):S46–50.

    PubMed  Google Scholar 

  8. Wan Y, Shang J, Sun S, Tai W, Chen J, Geng Q, He L, Chen Y, Wu J, Shi Z et al: Molecular mechanism for antibody-dependent enhancement of coronavirus entry. J Virol. 2020;94(5). https://pubmed.ncbi.nlm.nih.gov/31826992/.

  9. Liu L, Wei Q, Lin Q, Fang J, Wang H, Kwok H, Tang H, Nishiura K, Peng J, Tan Z et al: Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI insight. 2019;4(4). https://pubmed.ncbi.nlm.nih.gov/31826992/.

  10. Beigel JH, Aga E, Elie-Turenne MC, Cho J, Tebas P, Clark CL, Metcalf JP, Ozment C, Raviprakash K, Beeler J, et al. Anti-influenza immune plasma for the treatment of patients with severe influenza A: a randomised, double-blind, phase 3 trial. Lancet Respir Med. 2019;7(11):941–50.

    Article  Google Scholar 

  11. Dodd LE, Follmann D, Proschan M, Wang J, Malvy D, van Griensven J, Ciglenecki I, Horby PW, Ansumana R, Jiang JF, et al. A meta-analysis of clinical studies conducted during the West Africa Ebola virus disease outbreak confirms the need for randomized control groups. Sci Transl Med. 2019;11(520):eaaw1049. https://www.nature.com/articles/s41586-020-2355-0.

  12. Wu LP, Wang NC, Chang YH, Tian XY, Na DY, Zhang LY, Zheng L, Lan T, Wang LF, Liang GD. Duration of antibody responses after severe acute respiratory syndrome. Emerg Infect Dis. 2007;13(10):1562–4.

    Article  Google Scholar 

  13. Temperton NJ, Chan PK, Simmons G, Zambon MC, Tedder RS, Takeuchi Y, Weiss RA. Longitudinally profiling neutralizing antibody response to SARS coronavirus with pseudotypes. Emerg Infect Dis. 2005;11(3):411–6.

    Article  Google Scholar 

  14. Shen C, Wang Z, Zhao F, Yang Y, Li J, Yuan J, Wang F, Li D, Yang M, Xing L, et al. Treatment of 5 critically ill patients with COVID-19 with convalescent plasma. JAMA. 2020.

  15. Duan K, Liu B, Li C, Zhang H, Yu T, Qu J, Zhou M, Chen L, Meng S, Hu Y, et al. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc Natl Acad Sci U S A. 2020;117(17):9490–6.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Dr. Shankar-Hari is supported by the National Institute for Health Research (NIHR) Clinician Scientist Award (CS-2016-16-011). Prof. Menon is supported by an NIHR Senior Investigator award and through Cambridge NIHR Biomedical Research Centre funding. The views expressed in this publication are those of the author(s) and not necessarily those of the NHS, the National Institute for Health Research, or the Department of Health and Social Care. This manuscript is written on behalf of the United Kingdom SARS-CoV-2 Convalescent Plasma Evaluation (SCoPE) Consortium.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

MSH completed the first draft of the manuscript. LE, HH, DR, and DCM critically revised the manuscript for important intellectual content. All authors approved the final manuscript.

Corresponding author

Correspondence to Manu Shankar-Hari.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors are involved in randomised clinical trials of convalescent plasma in the UK. No other competing interests declared.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shankar-Hari, M., Estcourt, L., Harvala, H. et al. Convalescent plasma to treat critically ill patients with COVID-19: framing the need for randomised clinical trials. Crit Care 24, 449 (2020). https://doi.org/10.1186/s13054-020-03163-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13054-020-03163-3