Next Article in Journal
Modelling the Effect of MUC1 on Influenza Virus Infection Kinetics and Macrophage Dynamics
Next Article in Special Issue
Convalescent Plasma for Pregnant Women with COVID-19: A Systematic Literature Review
Previous Article in Journal
Multiplex PCR-Based Nanopore Sequencing and Epidemiological Surveillance of Hantaan orthohantavirus in Apodemus agrarius, Republic of Korea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Convalescent Plasma Transfusion for the Treatment of COVID-19 in Adults: A Global Perspective

College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
*
Author to whom correspondence should be addressed.
Submission received: 24 April 2021 / Revised: 2 May 2021 / Accepted: 5 May 2021 / Published: 7 May 2021

Abstract

:
More than one year into the novel coronavirus disease 2019 (COVID-19) pandemic, healthcare systems across the world continue to be overwhelmed with soaring daily cases. The treatment spectrum primarily includes ventilation support augmented with repurposed drugs and/or convalescent plasma transfusion (CPT) from recovered COVID-19 patients. Despite vaccine variants being recently developed and administered in several countries, challenges in global supply chain logistics limit their timely availability to the wider world population, particularly in developing countries. Given the measured success of conventional CPT in treating several infections over the past decade, recent studies have reported its effectiveness in decreasing the duration and severity of COVID-19 symptoms. In this review, we conduct a literature search of published studies investigating the use of CPT to treat COVID-19 patients from January 2020 to January 2021. The literature search identified 181 records of which 39 were included in this review. A random-effects model was used to aggregate data across studies, and mortality rates of 17 vs. 32% were estimated for the CPT and control patient groups, respectively, with an odds ratio (OR) of 0.49. The findings indicate that CPT shows potential in reducing the severity and duration of COVID-19 symptoms. However, early intervention (preferably within 3 days), recruitment of donors, and plasma potency introduce major challenges for its scaled-up implementation. Given the low number of existing randomized clinical trials (RCTs, four with a total of 319 patients), unanticipated risks to CPT recipients are highlighted and discussed. Nevertheless, CPT remains a promising COVID-19 therapeutic option that merits internationally coordinated RCTs to achieve a scientific risk–benefit consensus.

1. Introduction

Multiple cases of acute respiratory syndrome with unclear precursors were recorded during December 2019 in Wuhan, the capital of Hubei province in central China. Subsequent analysis of samples from patients’ lower respiratory tract indicated the presence of an unprecedented strain of human coronavirus, referred to as the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) [1,2,3], and its corresponding disease was dubbed coronavirus disease 2019 (COVID-19). Since the World Health Organization (WHO) declared the COVID-19 outbreak as a global pandemic on 11 March 2020, the cumulative number of global confirmed cases has surpassed 142 million with over 3 million deaths as of 21 April 2021 [4]. In response to growing pressures from the pandemic, more than 2000 clinical trials have been actively implemented during 2020, yet a unified treatment approach has not been recognized by the global healthcare and scientific communities to date [5]. Adding to the complexity of the current situation, government bodies and regulatory agencies overseeing national healthcare response plans have been subject to an unprecedented rate of trial reports published by multiple sources with often conflicting findings.
Global- and national-scale recovery timelines remain unclear amidst the uncertainty in treatment approaches and the emergence of new COVID-19 mutant strains [6]. Standard supportive care guidelines released by the WHO remain the status quo and mainly include symptom management through assisted ventilation and fluid management [7]. Remdesevir has been the only repurposed drug authorized by the United States (US) Food and Drug Administration (FDA) solely for emergency use [8]. Other drugs such as Hydroxychloroquinine [9], Ivermectin [10], and Oseltamivir [11] have been reportedly found to be comparably effective to standard supportive care for COVID-19 patients. Nevertheless, the generalized effectiveness of repurposed drugs remains subject to debate in the absence of large-scale data. As such, international efforts have been focused on vaccine development, which is naturally the product of several years of research and clinical trials before being authorized for general use.
The multi-phase vaccine development process encompasses pre-clinical testing (non-human trials), safety and dosage assessments (Phase 1), expanded safety trials (Phase 2), scalable efficacy trials (Phase 3), limited use authorization, and ultimately, full-scale approval. Given the urgency of the COVID-19 pandemic, a total of 89 vaccines are currently in human clinical trials—many of which are combining Phase 1 and 2 trials—while 27 have reached the final stages of testing [12]. Despite the timely progress in vaccine developments, two dynamic challenges hinder their post-efficacy stage for the global population. The primary set of challenges, especially for developing countries, lies in the supply chain and logistics in terms of scaled-up manufacturing and quality control, regional and local coordination of supply, and equality in distribution and governmental subsidies [13,14]. These factors exclusively stem from the post-efficacy stage, presumably after addressing any lurking uncertainties, such as the optimal timing and dosage of booster vaccine shots. Another transient challenge lies in the ongoing emergence of new SARS-CoV-2 variants [6,15]. The coronavirus genome is highly susceptible to mutations [16], which increases the likelihood of variant forms to evolve with time—a process termed genetic drift [17]. Hence, there is a sizable risk that a newly mutated virus might not be recognized by vaccine-acquired immune recognition from pre-mutation genomes.
Based on the above uncertainties in the COVID-19 response, numerous healthcare practitioners across the world have resorted to the conventional approach of convalescent plasma transfusion (CPT) therapy [18]. CPT therapy was first proposed in the late 1800s to treat a variety of previously unknown infectious diseases that lacked conclusive treatment options [19,20]. The general concept of CPT is based on the notion of relaying passive immunity by screening blood samples taken from a recovered individual (donor) for specific neutralizing antibodies, which are then administered to a patient suffering from the same disease to ameliorate symptoms and reduce mortality [21]. CPT has been particularly effective when used for diseases targeting the respiratory system, including the severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and influenza (H5N1 and H1N1) outbreaks [22].
Investigating the efficacy of CPT for treating COVID-19 patients has been recommended by the FDA since March 2020 [23]. Consequently, local-scale randomized clinical trials (RCTs), matched-control studies (MCSs), and case reports (CRs) conducted during 2020 offered preliminary evidence on the safety and efficacy of CPT therapy for COVID-19 patients [24]. However, the findings are subject to debate within the healthcare and scientific communities due to the lack of concrete evidence from statistically representative large-scale RCTs [25]. For instance, some studies were terminated early while reporting inconclusive results and a non-significant correlation between CPT and clinical improvement [26].
COVID-19 infections are generally mild and/or asymptomatic in the majority of the pediatric population with less than 2% of COVID-19 cases reported for infant and child age groups [27,28,29,30]. Additionally, current recommendations for the treatment of severe COVID-19 in children is limited to monitoring and supportive care [31]. Consequently, very few studies on convalescent plasma transfusion (CPT) therapy for COVID-19 pediatric patient groups are available to date. Rodriguez et al. [32] and Shankar et al. [33] provide case reports on individual high-risk patients with pre-existing conditions, namely, congenital heart disease (6 weeks old; female) and lymphoblastic leukemia (4 years old; female), respectively. In both cases, patients show a considerable reduction in symptoms after CPT therapy, which suggests a positive clinical correlation in infants and children with underlying conditions. Moreover, Figlerowicz et al. [34] reports the first attempt of CPT therapy on a 6-year-old child diagnosed with COVID-19-induced aplastic anemia without pre-existing conditions. Following the administration of antiviral drugs without signs of recovery during the first 5 weeks, a single dose of convalescent plasma (200 mL; 1:700 titer) resulted in recovery from COVID-19 within 3 weeks, while hematologic deficiencies persisted. The most representative attempt involving COVID-19 pediatric patients to date is the recent work of Małecki et al. [35]. Their study included a group of 13 patients (median age of 12 years), of which six CPT-recipient patients recovered within 3 days of transfusion, while the remaining seven control patients recovered after 12 days. Based on the limited data and a gap period of 10 days between infection and transfusion, the study suggests CPT may be a promising treatment for COVID-19 in children. Given the limited data on pediatric patients, underlying conditions, and/or notable lag times in administering CPT, the aforementioned studies were excluded from the review to avoid introducing significant biases within the pediatric age group.
The most recent reviews have been published before January 2021, including rapid and living systematic reviews and meta-analyses [25,36,37,38,39,40,41], after which several papers documenting trials on CPT as a treatment for COVID-19 have been published. Furthermore, previous reviews were largely based on limited RCT data. Taking into consideration the urgency of the topic and the struggle to identify treatments, it is vital that the literature is updated more often than in normal circumstances. Therefore, in this review, we conduct a global literature search of all studies examining the clinical effectiveness of CPT therapy for the treatment of COVID-19 and systematically analyze available data to date through a random-effects model. We also present a critical discussion on the limitations and challenges facing CPT therapy in the context of the ongoing COVID-19 response.

2. Materials and Methods

A comprehensive literature search was conducted by the first author (S.K.). The Cochrane Library, PubMed (MEDLINE), EMBASE, ScienceDirect, and Google Scholar were electronically searched from January 2020 until January 2021. The keywords related to COVID-19, CPT, and patients’ treatment concepts (“Convalescent”, “Plasma”, “Transfusion”, “COVID19”, “COVID-19”, “Coronavirus”, “SARS-CoV-2”, “Patient”, “Treatment”, “Passive”, and “Immunity”) were used with Boolean combinations.
No restriction on publication language was applied. In addition, the reference lists of articles resulting from the keyword search were manually inspected to identify additional relevant studies.

2.1. Eligibility Criteria

Studies were included in this review if they (1) are RCTs, MCSs, or CRs, which quantitatively report on CPT treatment for COVID-19 adult patients, (2) report trial data and performance metrics—in terms of the following parameters (at the minimum): number of patients, dosages, sampling periods, and mortality, and (3) report titers of SARS-CoV-2-neutralizing antibodies at a threshold therapeutic level of 1:80 [42,43]. Publications were excluded if they were conference proceedings, published as an abstract without the full text, or including patients under the age of 18 years old.

2.2. Data Synthesis

The clinical data were extracted and compiled from all studies and ingested into MATLAB® R2020a [44]. Boxplots for each study category (RCT, MCS, and CR) were produced using the Descriptive Statistics and Visualization toolbox in MATLAB [45]. Commonly used for descriptive statistics, boxplots graphically depict datasets through their quartile ranges, while outliers are represented as individual points. Importantly for our analysis, boxplots are non-parametric and avoid prescribing a statistical distribution to an unknown sample population. Hence, it is a standardized tool to inter-compare datasets based on their minimum, maximum, median, and interquartile ranges, with the spacing of the latter indicating the degree of variance in the data. Nevertheless, boxplots provide a coarse representation of the aggregated datasets and fail to account for inherent biases and uncertainties across studies, which may alter the findings. As such, a random-effects model was implemented in MATLAB® using the “LinearMixedModel” (LMM) embedded function [46], which returns estimates of the best linear unbiased predictors of random effects according to a prescribed significance level (5% used here). To ensure representative and generalized model output, a model ensemble was set up using three different methods for approximating the degrees of freedom (DOF), namely, the default constant definition, the Satterthwaite approximation, and the infinity definition [46,47].

3. Results

3.1. Included Studies

The literature search identified a total of 181 articles, of which 63 articles remained after removing duplicates. Following the title and abstract screening against the inclusion/exclusion criteria, 23 articles were excluded. During the full-text screening, one RCT article was excluded, since the majority of the trial’s CPT patients received plasma transfusions with non-detectable levels of SARS-CoV-2-neutralizing antibodies based on post-trial measurements (antibody titers < 1:80) [48]. The remaining 39 articles included in this review were geographically distributed across the following countries: China (13), USA (11), Mexico (2), South Korea (2), Turkey (2), Argentina (1), France (1), Hungary (1), Italy (1), Iran (1), Iraq (1), Netherlands (1), Qatar (1), and Spain (1).

3.2. Characteristics of Included Studies

The 39 studies included four RCTs [26,49,50,51], 14 MCSs [42,52,53,54,55,56,57,58,59,60,61,62,63,64], and 21 CRs [43,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84]. The majority of studies (28) were published in peer-reviewed journals, while 11 studies were available as pre-prints. Despite still being under peer-review, the raw data reported in these pre-prints remain highly relevant to our analysis, regardless of the methodology and recommendations provided.
An important uniformity across the studies was that all patients were included on the basis of demonstrating severe or life-threatening complications associated with COVID-19. Hence, all patients received one or more form of concomitant supportive care including mechanical ventilation and/or repurposed drugs as per WHO and local healthcare guidance. Almost half the studies (20) applied an antibody inclusion threshold to donor samples before transfusion, while the remaining (19) did not apply any screening to samples. The volume of transfused plasma ranged from 200 to 1200 mL, while the post-CPT therapy follow-up occurred between 7 and 60 days. Patient cohort age groups ranged from 23 to 96 years with a higher male proportion in each study. The total number of patients per study ranged from 49 to 103 for RCTs, from 20 to 3166 for MCSs, and from 1 to 31 for CRs. Table 1 lists the main characteristics relevant to our analysis and the results presented in the following section.

3.3. Statistical and Random-Effects Model Results

Figure 1 shows the boxplots of mortality rates as reported from the 39 studies and aggregated per RCTs, MCSs, and CRs with a total patient count of N = 319, 9655, and 170, respectively. The mortality rates for both the control and CPT recipient groups are shown, except for the CR studies which, by definition, are not assigned a control group. The median mortality rates of the control groups (RCT-Control and MCS-Control) are shown to be comparable at 24 and 28%, respectively. However, the MCS-Control group records a higher variance with mortality rates reaching around 45% compared to the 30% limit of the RCT-Control groups. Similarly, the median mortality rates of the corresponding CPT groups (RCT-CPT and MCS-CPT) are comparable at around 14%, while the MCS-CPT group shows a higher variance with mortality rates reaching 25%, compared to the 16% limit reported by the RCT-CPT group. The 28-fold difference in total patient numbers between the RCT (319) and MCS groups (9655) explains the higher variance in mortality rates from the latter group.
The results from both the RCT and MCS groups in Figure 1 indicate an overall reduction of 46% in mortality rates (~26 to 14%) in patients receiving CPT therapy compared to the control groups receiving standard supportive care and/or repurposed drugs. On the other hand, the CR-CPT group shows significantly lower mortality rates (less than 5%) compared to the RCT and MCS groups, but with outliers reaching 43%. This reflects the potential of high bias when aggregating individual case reports. For instance, several CR studies consisted of COVID-19 patients with impaired immunities, which suppressed their response to standard supportive care [68,79,83]. However, their symptoms were significantly reduced and showed signs of recovery after 3–5 days of CPT with antibody titer levels higher than 1:80, compared to undetectable levels pre-CPT.
To address the risk of bias and generalize the boxplot results, a random-effects model was set up based on the parameters listed in Table 1. Mortality rates were derived based on the longest follow-up period in each study, and odds ratios (ORs) were aggregated for each of the RCT and MCS study categories (CRs do not have control groups). A meta-regression analysis based on the MATLAB LMM function [46] was used to evaluate the significance of the parameters listed in Table 1 (i.e., cohort age, follow-up duration, plasma volume/dose) with respect to mortality across the studies. Hypothesis testing using the two-tailed test at a 95% confidence interval (5% significance) was carried out.
The random-effects analysis showed median mortality rates of 11 vs. 23% for the CPT and control groups, respectively, across the four RCT studies with an OR of 0.41. This suggests that CPT was correlated with a 59% reduction in the likelihood of mortality among COVID-19 patients. This association was further observed based on the MCS random-effects model results, for which mortality rates of 21 vs. 32% for the CPT and control groups with an OR of 0.47 (i.e., 53% reduction in the likelihood of mortality). To further investigate these findings, an overall random-effects model was used to aggregate results from both RCT and MC studies. Similarly, mortality rates of 17 vs. 32% for the CPT and control groups were recorded, respectively, with an OR of 0.49—a 51% reduction in the overall likelihood of mortality across all trials. The meta-regression analyses indicated that cohort age and the follow-up duration did not affect the aggregate OR computed for all clinical trials.

4. Discussion

The FDA clearly states that the authorization of CPT for emergency use is not meant to replace ongoing clinical trials, since there is insufficient evidence to currently designate CPT as a new standard of care [23]. In this review, a total of 39 trials and case reports were analyzed and systematically inter-compared to assess the efficacy of CPT for the treatment of COVID-19 patients. Overall, the findings of this review support the efficacy of CPT therapy for the treatment of COVID-19 patients, particularly those experiencing severe symptoms. This outcome generally contests the findings of former systematic reviews on the efficacy of CPT therapy for COVID-19, with some reporting inconclusive results [26,37]. Nevertheless, several underlying risks and limitations may undermine the efficacy of CPT therapy in the context of COVID-19.

4.1. Plasma Potency: SARS-CoV-2 Antibody Inclusion Thresholds

As the primary active agents in convalescent plasma, neutralizing antibodies are considered the key indicator of plasma potency for CPT therapy. However, only two of the 39 studies explicitly reported a significant correlation between mortality and SARS-CoV-2-neutralizing antibody titer levels in CPT doses. Interestingly, despite the importance of verifying plasma potency, almost half the studies (18) did not screen donor plasma samples to ensure adequate levels of SARS-CoV-2 antibodies [49,53,54,55,60,62,63,67,69,70,72,74,75,76,80,81,82,84]. In one RCT trial in India by Agarwal et al. [48], the majority of patients (71%) received CPT doses with relatively low SARS-CoV-2-neutralizing antibody titer (<1:80), while the remaining patients received non-detectable levels. There is a consensus among trials to date that an adequate titer of neutralizing antibodies should exceed 1:80 [42,43]. Hence, the Agarwal et al. [48] RCT was excluded from the current review (and Figure 1), as it lacked the fundamental biological and therapeutic mechanism of CPT.
It is important to note that different serologic tests were used to report antibody levels across the 20 studies, which performed plasma screening (see Table 1). The different assay results included the enzyme-linked immunosorbent assay (ELSA) and neutralizing antibody (NA) indices (i) or titers (t), signal-to-cutoff ratio (S/Co), and total antibodies (TA) in arbitrary units per volume (AU/mL). Several types of assays have been developed in a short period of time in response to the COVID-19 outbreak, each with its own set of uncertainties in terms of clinical sensitivity and specificity, such as sample collection and handling, technological challenges, unknown systematic error margins, and dependencies on patient health conditions [85]. According to the most recent literature, a single assay result will not reflect a representative quantitative measure, whereas merging different test results can help minimize biases and provide more conclusive therapeutic thresholds [86,87].

4.2. CPT Intervention Time

In addition to plasma potency and the need for uniform screening approaches, early intervention is an equally significant factor to ensure CPT efficacy in treating COVID-19 patients. Joyner et al. [64] documented results from an Expanded Access Program for the treatment of COVID-19 patients with CPT. The study consisted of 35,322 patients spanning across 2807 acute care facilities in the US. While the program did not include a control group, results showed that transfusions within three days of COVID-19 diagnosis yielded an average reduction of 5% in mortality rates compared to those receiving transfusions after 4 or more days of diagnosis. Additionally, the trial by Zeng et al. [55] recorded the highest mortality rates across all studies (see Table 1) for both the CPT recipient (83%) and control (93%) groups. This study also reported the longest time for intervention compared to all existing trials—more than 21 days after diagnosis. Hence, CPT is generally not effective in reducing mortality in critically ill patients and is directly linked to the scalability of testing and contact tracing to ensure early intervention.

4.3. Donor–Recipient Risks

Among the reviewed articles, only Duan et al. [42] reported mild CPT side effects. However, previous studies on CPT therapy in patients infected by diseases other than COVID-19 have reported adverse (and occasionally severe) reactions. In general, the risk of a thrombotic event in CPT recipients can reach up to 15% [88]. More specifically, Luke et al. [19] conducted CPT trials on Spanish influenza (H5N1) patients who experienced tremors, facial spotting, fever, and transfusion-associated circulatory overload (TACO) [89]. Based on CPT trials on Ebola patients by Takada et al. [90], the transmitted antibodies caused complement binding to the virus, which strengthened the infection in recipient cells. Further research is needed to rule out the likelihood of this complement effect in COVID-19 patients. In terms of donor safety, CPT may cause mild and short-lived side effects, including dizziness, fatigue, and fainting associated with the loss of water and nutrients through the plasma extraction. Other CPT process-based inadequacies in terms of sterility or expertise may lead to bacterial or viral transmissions to a donor and, in less common cases, a fatal citrate reaction, which may trigger cardiac arrest [91].

4.4. Importance of Large-Scale RCTs

Despite the preliminary evidence on the efficacy of CPT for treating hospitalized COVID-19 patients, the lack of sufficient RCT data remains a critical issue. Many researchers refer to unsatisfactory results from RCTs on CPT in influenza A [92,93] and respiratory syncytial virus (RSV) [94] patients. While large-scale RCTs are understandably challenging given the relatively short duration since the emergence of COVID-19, they remain instrumental to reaching a scientific consensus regarding the efficacy of CPT. However, scalable RCTs warrant considerable investment and resources, which may be diverted away from other areas of the pandemic response [89]. For instance, testing large numbers of CPT donors as part of their screening process may reduce the rate of public health testing for contact tracing and early intervention. Hence, a careful optimization approach based on mathematical modeling is recommended to assess the magnitude of this resource diversion to scalable RCTs and the overall tradeoff.
An important limitation to the review is the merging of mortality rates across studies with different patient group sizes, in addition to biases from differences in the nation of study origin, concomitant treatment approaches, antibody titer, dosage volume, and duration of post-CPT patient follow-up. However, given the shortage in large-scale RCT data, the aggregation of data from multiple CPT trial formats provides important insights for both its preliminary efficacy and for the design of more representative and scalable RCTs.

5. Conclusions

Aggregated results show an overall reduction of 46% in mortality in patients receiving CPT therapy compared to control groups receiving standard supportive care and/or repurposed drugs. Despite the promising preliminary evidence from trials and case studies to date, several challenges remain in terms of the technical, safety, and resource aspects of CPT trials as presented in Section 4. Early intervention is shown to be a critical factor for CPT efficacy in treating COVID-19 patients, which is coupled to the scalability of testing and contact tracing. The recruitment of plasma donors, consistency of adequate ranges of SARS-CoV-2 antibody titer, risks of adverse effects to CPT donors/recipients, and robust management of healthcare resources are some of the major challenges to consider before implementing large-scale RCTs.

Author Contributions

Conceptualization, S.K. and B.A.-O.; methodology, S.K.; validation, B.A.-O.; formal analysis, S.K.; investigation, S.K.; data curation, S.K.; writing—original draft preparation, S.K.; writing—review and editing, B.A.-O.; supervision, B.A.-O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

All data incorporated in this review are presented and duly cited in Table 1.

Acknowledgments

The authors thank Abdu Adem and Abderrahim Oulhaj for providing feedback on the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dong, E.; Du, H.; Gardner, L. An interactive web-based dashboard to track COVID-19 in real time. Lancet Infect. Dis. 2020, 20, 533–534. [Google Scholar] [CrossRef]
  2. Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [Green Version]
  3. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.-M.; Wang, W.; Song, Z.-G.; Hu, Y.; Tao, Z.-W.; Tian, J.-H.; Pei, Y.-Y. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 265–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. WHO. Coronavirus (COVID-19). Available online: https://who.sprinklr.com/ (accessed on 26 January 2021).
  5. Siemieniuk, R.A.; Bartoszko, J.J.; Ge, L.; Zeraatkar, D.; Izcovich, A.; Kum, E.; Pardo-Hernandez, H.; Rochwerg, B.; Lamontagne, F.; Han, M.A. Drug treatments for covid-19: Living systematic review and network meta-analysis. BMJ 2020, 370, m2980. [Google Scholar] [CrossRef] [PubMed]
  6. Van Oosterhout, C.; Hall, N.; Ly, H.; Tyler, K.M. COVID-19 Evolution during the Pandemic–Implications of New SARS-CoV-2 Variants on Disease Control and Public Health Policies; Taylor & Francis: Milton Park/Abingdon, UK, 2021. [Google Scholar]
  7. WHO. Clinical Management of Severe Acute Respiratory Infections (SARI) When COVID-19 Disease Is Suspected; Interim Guidance; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  8. FDA. Coronavirus (COVID-19) Update: FDA Issues Emergency Use Authorization for Potential COVID-19 Treatment; Food and Drug Administration: Silver Spring, MD, USA, 2020. [Google Scholar]
  9. Gautret, P.; Lagier, J.-C.; Parola, P.; Meddeb, L.; Mailhe, M.; Doudier, B.; Courjon, J.; Giordanengo, V.; Vieira, V.E.; Dupont, H.T. Hydroxychloroquine and azithromycin as a treatment of COVID-19: Results of an open-label non-randomized clinical trial. Int. J. Antimicrob. Agents 2020, 56, 105949. [Google Scholar] [CrossRef] [PubMed]
  10. Caly, L.; Druce, J.D.; Catton, M.G.; Jans, D.A.; Wagstaff, K.M. The FDA-approved drug ivermectin inhibits the replication of SARS-CoV-2 in vitro. Antivir. Res. 2020, 178, 104787. [Google Scholar] [CrossRef] [PubMed]
  11. Cunningham, A.C.; Goh, H.P.; Koh, D. Treatment of COVID-19: Old Tricks for New Challenges; Springer: Berlin/Heidelberg, Germany, 2020. [Google Scholar]
  12. Corum, J.; Grady, D.; Wee, S.-L.; Zimmer, C. Coronavirus Vaccine Tracker; The New York Times: New York, NY, USA, 2020; p. 5. [Google Scholar]
  13. Kim, J.H.; Marks, F.; Clemens, J.D. Looking beyond COVID-19 vaccine phase 3 trials. Nat. Med. 2021, 27, 205–211. [Google Scholar] [CrossRef]
  14. Newton, P.N.; Bond, K.C.; Adeyeye, M.; Antignac, M.; Ashenef, A.; Awab, G.R.; Bannenberg, W.J.; Bower, J.; Breman, J.; Brock, A. COVID-19 and risks to the supply and quality of tests, drugs, and vaccines. Lancet Glob. Health 2020, 8, e754–e755. [Google Scholar] [CrossRef]
  15. Koyama, T.; Weeraratne, D.; Snowdon, J.L.; Parida, L. Emergence of drift variants that may affect COVID-19 vaccine development and antibody treatment. Pathogens 2020, 9, 324. [Google Scholar] [CrossRef] [PubMed]
  16. Yao, H.-P.; Lu, X.; Chen, Q.; Xu, K.; Chen, Y.; Cheng, L.; Liu, F.; Wu, Z.; Wu, H.; Jin, C. Patient-Derived Mutations Impact Pathogenicity of SARS-CoV-2. CELL-D-20-01124. 2020. Available online: https://www.medrxiv.org/content/10.1101/2020.04.14.20060160v2 (accessed on 6 May 2021).
  17. Lynch, M.; Ackerman, M.S.; Gout, J.-F.; Long, H.; Sung, W.; Thomas, W.K.; Foster, P.L. Genetic drift, selection and the evolution of the mutation rate. Nat. Rev. Genet. 2016, 17, 704. [Google Scholar] [CrossRef]
  18. Chen, L.; Xiong, J.; Bao, L.; Shi, Y. Convalescent plasma as a potential therapy for COVID-19. Lancet Infect. Dis. 2020, 20, 398–400. [Google Scholar] [CrossRef]
  19. Luke, T.C.; Kilbane, E.M.; Jackson, J.L.; Hoffman, S.L. Meta-analysis: Convalescent blood products for Spanish influenza pneumonia: A future H5N1 treatment? Ann. Intern. Med. 2006, 145, 599–609. [Google Scholar] [CrossRef] [PubMed]
  20. Marano, G.; Vaglio, S.; Pupella, S.; Facco, G.; Catalano, L.; Liumbruno, G.M.; Grazzini, G. Convalescent plasma: New evidence for an old therapeutic tool? Blood Transfus. 2016, 14, 152. [Google Scholar] [PubMed] [Green Version]
  21. Bozzo, J.; Jorquera, J.I. Use of human immunoglobulins as an anti-infective treatment: The experience so far and their possible re-emerging role. Expert Rev. Anti-Infect. Ther. 2017, 15, 585–604. [Google Scholar] [CrossRef]
  22. Mair-Jenkins, J.; Saavedra-Campos, M.; Baillie, J.K.; Cleary, P.; Khaw, F.-M.; Lim, W.S.; Makki, S.; Rooney, K.D.; Group, C.P.S.; Nguyen-Van-Tam, J.S. The effectiveness of convalescent plasma and hyperimmune immunoglobulin for the treatment of severe acute respiratory infections of viral etiology: A systematic review and exploratory meta-analysis. J. Infect. Dis. 2015, 211, 80–90. [Google Scholar] [CrossRef] [Green Version]
  23. FDA. Recommendations for Investigational COVID-19 Convalescent Plasma, Content Current as of: 04/13/2020; Food and Drug Administration: Silver Spring, MD, USA, 2020. [Google Scholar]
  24. Joyner, M.J.; Klassen, S.A.; Senefeld, J.; Johnson, P.W.; Carter, R.E.; Wiggins, C.C.; Shoham, S.; Grossman, B.J.; Henderson, J.P.; Musser, J.M. Evidence favouring the efficacy of convalescent plasma for COVID-19 therapy. medRxiv 2020. [Google Scholar] [CrossRef]
  25. Valk, S.J.; Piechotta, V.; Chai, K.L.; Doree, C.; Monsef, I.; Wood, E.M.; Lamikanra, A.; Kimber, C.; McQuilten, Z.; So-Osman, C. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: A rapid review. Cochrane Database Syst. Rev. 2020. [Google Scholar] [CrossRef]
  26. Li, L.; Zhang, W.; Hu, Y.; Tong, X.; Zheng, S.; Yang, J.; Kong, Y.; Ren, L.; Wei, Q.; Mei, H. 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, 324, 460–470. [Google Scholar] [CrossRef]
  27. Dong, Y.; Mo, X.; Hu, Y.; Qi, X.; Jiang, F.; Jiang, Z.; Tong, S. Epidemiology of COVID-19 among children in China. Pediatrics 2020, 145, e20200702. [Google Scholar] [CrossRef] [Green Version]
  28. DeBiasi, R.L.; Song, X.; Delaney, M.; Bell, M.; Smith, K.; Pershad, J.; Ansusinha, E.; Hahn, A.; Hamdy, R.; Harik, N. Severe coronavirus disease-2019 in children and young adults in the Washington, DC, metropolitan region. J. Pediatr. 2020, 223, 199–203.e1. [Google Scholar] [CrossRef]
  29. Zimmermann, P.; Curtis, N. Coronavirus infections in children including COVID-19: An overview of the epidemiology, clinical features, diagnosis, treatment and prevention options in children. Pediatr. Infect. Dis. J. 2020, 39, 355. [Google Scholar] [CrossRef] [PubMed]
  30. Ludvigsson, J.F. Systematic review of COVID-19 in children shows milder cases and a better prognosis than adults. Acta Paediatr. 2020, 109, 1088–1095. [Google Scholar] [CrossRef]
  31. Ye, Z.; Rochwerg, B.; Wang, Y.; Adhikari, N.K.; Murthy, S.; Lamontagne, F.; Fowler, R.A.; Qiu, H.; Wei, L.; Sang, L. Treatment of patients with nonsevere and severe coronavirus disease 2019: An evidence-based guideline. CMAJ 2020, 192, E536–E545. [Google Scholar] [CrossRef]
  32. Rodriguez, Z.; Shane, A.L.; Verkerke, H.; Lough, C.; Zimmerman, M.G.; Suthar, M.; Wrammert, J.; MacDonald, H.; Wolf, M.; Clarke, S. COVID-19 convalescent plasma clears SARS-CoV-2 refractory to remdesivir in an infant with congenital heart disease. Blood Adv. 2020, 4, 4278. [Google Scholar] [CrossRef]
  33. Shankar, R.; Radhakrishnan, N.; Dua, S.; Arora, S.; Rana, M.; Sahu, D.K.; Rai, S.; Gupta, D.K. Convalescent plasma to aid in recovery of COVID-19 pneumonia in a child with acute lymphoblastic leukemia. Transfus. Apher. Sci. 2021, 60, 102956. [Google Scholar] [CrossRef]
  34. Figlerowicz, M.; Mania, A.; Lubarski, K.; Lewandowska, Z.; Służewski, W.; Derwich, K.; Wachowiak, J.; Mazur-Melewska, K. First case of convalescent plasma transfusion in a child with COVID-19-associated severe aplastic anemia. Transfus. Apher. Sci. 2020, 59, 102866. [Google Scholar] [CrossRef]
  35. Małecki, P.; Faltin, K.; Mania, A.; Mazur-Melewska, K.; Cwalińska, A.; Zawadzka, A.; Bukowska, A.; Lisowska, K.; Graniczna, K.; Figlerowicz, M. Effects and Safety of Convalescent Plasma Administration in a Group of Polish Pediatric Patients with COVID-19: A Case Series. Life 2021, 11, 247. [Google Scholar] [CrossRef]
  36. Gempeler, A.; Rada, G.; Rojas, L. Convalescent Plasma for the Treatment of Patients with COVID-19: A Living Systematic Review. PROSPERO 2020 CRD42020180703. Available online: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42020180703 (accessed on 6 May 2021).
  37. Chai, K.L.; Valk, S.J.; Piechotta, V.; Kimber, C.; Monsef, I.; Doree, C.; Wood, E.M.; Lamikanra, A.A.; Roberts, D.J.; McQuilten, Z. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: A living systematic review. Cochrane Database Syst. Rev. 2020. [Google Scholar] [CrossRef]
  38. Rajendran, K.; Krishnasamy, N.; Rangarajan, J.; Rathinam, J.; Natarajan, M.; Ramachandran, A. Convalescent plasma transfusion for the treatment of COVID-19: Systematic review. J. Med. Virol. 2020, 92, 1475–1483. [Google Scholar] [CrossRef]
  39. Wang, Y.; Huo, P.; Dai, R.; Lv, X.; Yuan, S.; Zhang, Y.; Guo, Y.; Li, R.; Yu, Q.; Zhu, K. Convalescent plasma may be a possible treatment for COVID-19: A systematic review. Int. Immunopharmacol. 2021, 91, 107262. [Google Scholar] [CrossRef]
  40. Devarasetti, P.K.; Rajasekhar, L.; Baisya, R.; Sreejitha, K.; Vardhan, Y.K. A review of COVID-19 convalescent plasma use in COVID-19 with focus on proof of efficacy. Immunol. Res. 2021, 69, 18–25. [Google Scholar] [CrossRef]
  41. Al-Riyami, A.Z.; Schäfer, R.; van den Berg, K.; Bloch, E.M.; Estcourt, L.J.; Goel, R.; Hindawi, S.; Josephson, C.D.; Land, K.; McQuilten, Z.K. Clinical use of convalescent plasma in the covid-19 pandemic: A transfusion-focussed gap analysis with recommendations for future research priorities. Vox Sang. 2021, 116, 88–98. [Google Scholar] [CrossRef] [PubMed]
  42. Duan, K.; Liu, B.; Li, C.; Zhang, H.; Yu, T.; Qu, J.; Zhou, M.; Chen, L.; Meng, S.; Hu, Y. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc. Natl. Acad. Sci. USA 2020, 117, 9490–9496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Shen, C.; Wang, Z.; Zhao, F.; Yang, Y.; Li, J.; Yuan, J.; Wang, F.; Li, D.; Yang, M.; Xing, L. Treatment of 5 critically ill patients with COVID-19 with convalescent plasma. JAMA 2020, 323, 1582–1589. [Google Scholar] [CrossRef]
  44. MATLAB Version 9.8.0 (R2020a); The MathWorks Inc.: Natick, MA, USA, 2020.
  45. Martinez, W.L.; Martinez, A.R.; Solka, J. Exploratory Data Analysis with MATLAB; CRC Press: Boca Raton, FL, USA, 2017. [Google Scholar]
  46. Worsley, K.J.; Taylor, J.; Carbonell, F.; Chung, M.; Duerden, E.; Bernhardt, B.; Lyttelton, O.; Boucher, M.; Evans, A. A Matlab toolbox for the statistical analysis of univariate and multivariate surface and volumetric data using linear mixed effects models and random field theory. NeuroImage 2009, 47, S102. [Google Scholar] [CrossRef]
  47. Satterthwaite, F.E. An approximate distribution of estimates of variance components. Biom. Bull. 1946, 2, 110–114. [Google Scholar] [CrossRef]
  48. Agarwal, A.; Mukherjee, A.; Kumar, G.; Chatterjee, P.; Bhatnagar, T.; Malhotra, P. Convalescent plasma in the management of moderate covid-19 in adults in India: Open label phase II multicentre randomised controlled trial (PLACID Trial). BMJ 2020, 371, m3939. [Google Scholar] [CrossRef]
  49. Avendano-Sola, C.; Ramos-Martinez, A.; Munez-Rubio, E.; Ruiz-Antoran, B.; de Molina, R.M.; Torres, F.; Fernandez-Cruz, A.; Callejas-Diaz, A.; Calderon, J.; Payares-Herrera, C. Convalescent Plasma for COVID-19: A multicenter, randomized clinical trial. medRxiv 2020. [Google Scholar] [CrossRef]
  50. Rasheed, A.M.; Fatak, D.F.; Hashim, H.A.; Maulood, M.F.; Kabah, K.K.; Abdulamir, A.S. The therapeutic potential of convalescent plasma therapy on treating critically-ill COVID-19 patients residing in respiratory care units in hospitals in Baghdad, Iraq. medRxiv 2020. [Google Scholar] [CrossRef]
  51. Gharbharan, A.; Jordans, C.C.; GeurtsvanKessel, C.; den Hollander, J.G.; Karim, F.; Mollema, F.P.; Stalenhoef, J.E.; Dofferhoff, A.; Ludwig, I.; Koster, A. Convalescent Plasma for COVID-19. A randomized clinical trial. medRxiv 2020. [Google Scholar] [CrossRef]
  52. Perotti, C.; Baldanti, F.; Bruno, R.; Del Fante, C.; Seminari, E.; Casari, S.; Percivalle, E.; Glingani, C.; Musella, V.; Belliato, M. Mortality reduction in 46 patients with severe COVID-19 treated with hyperimmune plasma. A proof-of-concept, single-arm, multicenter trial. Haematologica 2020, 105, 2834–2840. [Google Scholar] [CrossRef] [PubMed]
  53. Omrani, A.S.; Zaqout, A.; Baiou, A.; Daghfal, J.; Elkum, N.; Alattar, R.A.; Bakdach, D.; Abusriwil, H.; Mostafa, A.M.; Alhariri, B. Convalescent plasma for the treatment of patients with severe coronavirus disease 2019: A preliminary report. J. Med. Virol. 2021, 93, 1678–1686. [Google Scholar] [CrossRef]
  54. Hegerova, L.; Gooley, T.A.; Sweerus, K.A.; Maree, C.; Bailey, N.; Bailey, M.; Dunleavy, V.; Patel, K.; Alcorn, K.; Haley, R. Use of convalescent plasma in hospitalized patients with COVID-19: Case series. Blood J. Am. Soc. Hematol. 2020, 136, 759–762. [Google Scholar] [CrossRef]
  55. Zeng, Q.-L.; Yu, Z.-J.; Gou, J.-J.; Li, G.-M.; Ma, S.-H.; Zhang, G.-F.; Xu, J.-H.; Lin, W.-B.; Cui, G.-L.; Zhang, M.-M. Effect of convalescent plasma therapy on viral shedding and survival in patients with coronavirus disease 2019. J. Infect. Dis. 2020, 222, 38–43. [Google Scholar] [CrossRef] [PubMed]
  56. Donato, M.; Park, S.; Baker, M.; Korngold, R.; Morawski, A.; Geng, X.; Tan, M.T.; Rowley, S.; Chow, K.; Brown, E. Clinical and laboratory evaluation of patients with SARS-CoV-2 pneumonia treated with high-titer convalescent plasma: A prospective study. medRxiv 2020. [Google Scholar] [CrossRef]
  57. Salazar, M.R.; Gonzalez, S.E.; Regairaz, L.; Ferrando, N.S.; Gonzalez, V.; Carrera, P.M.; Muñoz, L.; Pesci, S.A.; Vidal, J.M.; Kreplak, N. Effect of convalescent plasma on mortality in patients with COVID-19 pneumonia. medRxiv 2020. [Google Scholar] [CrossRef]
  58. Salazar, E.; Christensen, P.A.; Graviss, E.A.; Ngyuen, D.T.; Castillo, B.; Chen, J.; Lopez, B.V.; Eager, T.; Yi, X.; Zhao, P. Early transfusion of a large cohort of COVID-19 patients with high titer anti-SARS-CoV-2 spike protein IgG convalescent plasma confirms a signal of significantly decreased mortality. medRxiv 2020. [Google Scholar] [CrossRef]
  59. Liu, S.T.; Lin, H.-M.; Baine, I.; Wajnberg, A.; Gumprecht, J.P.; Rahman, F.; Rodriguez, D.; Tandon, P.; Bassily-Marcus, A.; Bander, J. Convalescent plasma treatment of severe COVID-19: A propensity score–matched control study. Nat. Med. 2020, 26, 1708–1713. [Google Scholar] [CrossRef]
  60. Xia, X.; Li, K.; Wu, L.; Wang, Z.; Zhu, M.; Huang, B.; Li, J.; Wang, Z.; Wu, W.; Wu, M. Improved clinical symptoms and mortality on severe/critical COVID-19 patients utilizing convalescent plasma transfusion. Blood 2020, 136, 755–759. [Google Scholar] [CrossRef]
  61. Abolghasemi, H.; Eshghi, P.; Cheraghali, A.M.; Fooladi, A.A.I.; Moghaddam, F.B.; Imanizadeh, S.; Maleki, M.M.; Ranjkesh, M.; Rezapour, M.; Bahramifar, A. Clinical efficacy of convalescent plasma for treatment of COVID-19 infections: Results of a multicenter clinical study. Transfus. Apher. Sci. 2020, 59, 102875. [Google Scholar] [CrossRef] [PubMed]
  62. Rogers, R.; Shehadeh, F.; Mylona, E.; Rich, J.; Neill, M.; Touzard-Romo, F.; Geffert, S.; Larkin, J.; Bailey, J.A.; Lu, S. Convalescent plasma for patients with severe COVID-19: A matched cohort study. medRxiv 2020. [Google Scholar] [CrossRef] [PubMed]
  63. Altuntas, F.; Ata, N.; Yigenoglu, T.N.; Bascı, S.; Dal, M.S.; Korkmaz, S.; Namdaroglu, S.; Basturk, A.; Hacıbekiroglu, T.; Dogu, M.H. Convalescent plasma therapy in patients with COVID-19. Transfus. Apher. Sci. 2020, 60, 102955. [Google Scholar] [CrossRef]
  64. Joyner, M.J.; Senefeld, J.W.; Klassen, S.A.; Mills, J.R.; Johnson, P.W.; Theel, E.S.; Wiggins, C.C.; Bruno, K.A.; Klompas, A.M.; Lesser, E.R. Effect of convalescent plasma on mortality among hospitalized patients with COVID-19: Initial three-month experience. medRxiv 2020. [Google Scholar] [CrossRef]
  65. Chen, S.; Lu, C.; Li, P.; Wang, L.; Wang, H.; Yang, Q.; Chen, L.; Li, J.; Ma, H.; Sang, Q. Effectiveness of Convalescent Plasma for Treatment of COVID-19 Patients. medRxiv 2020. [Google Scholar] [CrossRef]
  66. Martinez-Resendez, M.F.; Castilleja-Leal, F.; Torres-Quintanilla, A.; Rojas-Martinez, A.; Garcia-Rivas, G.; Ortiz-Lopez, R.; Trevino, V.; Lara-Medrano, R.; Villanueva-Lozano, H.; Ramirez-Elizondo, T. Initial experience in Mexico with convalescent plasma in COVID-19 patients with severe respiratory failure, a retrospective case series. medRxiv 2020. [Google Scholar] [CrossRef]
  67. Jin, C.; Gu, J.; Yuan, Y.; Long, Q.; Zhang, Q.; Zhou, H.; Wu, W.; Zhang, W. Treatment of 6 COVID-19 patients with convalescent plasma. medRxiv 2020. [Google Scholar] [CrossRef]
  68. Jin, H.; Reed, J.C.; Liu, S.T.; Ho, H.-E.; Lopes, J.P.; Ramsey, N.B.; Waqar, O.; Rahman, F.; Aberg, J.A.; Bouvier, N.M. Three patients with X-linked agammaglobulinemia hospitalized for COVID-19 improved with convalescent plasma. J. Allergy Clin. Immunol. Pract. 2020, 8, 3594–3596.e3. [Google Scholar] [CrossRef] [PubMed]
  69. Zhang, B.; Liu, S.; Tan, T.; Huang, W.; Dong, Y.; Chen, L.; Chen, Q.; Zhang, L.; Zhong, Q.; Zhang, X. Treatment with convalescent plasma for critically ill patients with severe acute respiratory syndrome coronavirus 2 infection. Chest 2020, 158, e9–e13. [Google Scholar] [CrossRef] [PubMed]
  70. Ahn, J.Y.; Sohn, Y.; Lee, S.H.; Cho, Y.; Hyun, J.H.; Baek, Y.J.; Jeong, S.J.; Kim, J.H.; Ku, N.S.; Yeom, J.-S. Use of convalescent plasma therapy in two COVID-19 patients with acute respiratory distress syndrome in Korea. J. Korean Med. Sci. 2020, 35, e149. [Google Scholar] [CrossRef] [Green Version]
  71. Bobek, I.; Gopcsa, L.; Réti, M.; Bekő, G.; Hancz, L.; Lakatos, B.; Molnár, E.; Nagy, S.; Reményi, P.; Sebestyén, G. Successful administration of convalescent plasma in critically ill COVID-19 patients in Hungary: The first two cases. Orv. Hetil. 2020, 161, 1111–1121. [Google Scholar] [CrossRef]
  72. Im, J.H.; Nahm, C.H.; Baek, J.H.; Kwon, H.Y.; Lee, J.-S. Convalescent plasma therapy in coronavirus disease 2019: A case report and suggestions to overcome obstacles. J. Korean Med. Sci. 2020, 35, e239. [Google Scholar] [CrossRef] [PubMed]
  73. Peng, H.; Gong, T.; Huang, X.; Sun, X.; Luo, H.; Wang, W.; Luo, J.; Luo, B.; Chen, Y.; Wang, X. A synergistic role of convalescent plasma and mesenchymal stem cells in the treatment of severely ill COVID-19 patients: A clinical case report. Stem Cell Res. Ther. 2020, 11, 291. [Google Scholar] [CrossRef] [PubMed]
  74. Xu, T.-M.; Lin, B.; Chen, C.; Liu, L.-G.; Xue, Y. Non-optimal effectiveness of convalescent plasma transfusion and hydroxychloroquine in treating COVID-19: A case report. Virol. J. 2020, 17, 80. [Google Scholar] [CrossRef]
  75. Anderson, J.; Schauer, J.; Bryant, S.; Graves, C.R. The use of convalescent plasma therapy and remdesivir in the successful management of a critically ill obstetric patient with novel coronavirus 2019 infection: A case report. Case Rep. Women’s Health 2020, 27, e00221. [Google Scholar] [CrossRef]
  76. Bao, Y.; Lin, S.Y.; Cheng, Z.H.; Xia, J.; Sun, Y.P.; Zhao, Q.; Liu, G.J. Clinical features of COVID-19 in a young man with massive cerebral hemorrhage—Case report. SN Compr. Clin. Med. 2020, 2, 703–709. [Google Scholar] [CrossRef] [PubMed]
  77. Çınar, O.E.; Sayınalp, B.; Karakulak, E.A.; Karataş, A.A.; Velet, M.; İnkaya, A.Ç.; Ortaç, N.E.E.; Öcal, S.; Aksu, S.; Haznedaroğlu, İ.C. Convalescent (immune) plasma treatment in a myelodysplastic COVID-19 patient with disseminated tuberculosis. Transfus. Apher. Sci. 2020, 59, 102821. [Google Scholar] [CrossRef]
  78. Kong, Y.; Cai, C.; Ling, L.; Zeng, L.; Wu, M.; Wu, Y.; Zhang, W.; Liu, Z. Successful treatment of a centenarian with coronavirus disease 2019 (COVID-19) using convalescent plasma. Transfus. Apher. Sci. 2020, 59, 102820. [Google Scholar] [CrossRef] [PubMed]
  79. Hueso, T.; Pouderoux, C.; Péré, H.; Beaumont, A.-L.; Raillon, L.-A.; Ader, F.; Chatenoud, L.; Eshagh, D.; Szwebel, T.-A.; Martinot, M. Convalescent plasma therapy for B-cell–depleted patients with protracted COVID-19. Blood J. Am. Soc. Hematol. 2020, 136, 2290–2295. [Google Scholar]
  80. Hartman, W.R.; Hess, A.S.; Connor, J.P. Hospitalized COVID-19 patients treated with Convalescent Plasma in a mid-size city in the midwest. Transl. Med. Commun. 2020, 5, 17. [Google Scholar] [CrossRef]
  81. Jamous, F.; Meyer, N.; Buus, D.; Ateeli, H.; Taggart, K.; Devasahayam, J.; Hanson, T.; Alzoubaidi, M.; Nazir, J. Critical Illness Due to Covid-19: A Description of the Surge in a Single Center in Sioux Falls. South Dak. Med. 2020, 73, 312–317. [Google Scholar]
  82. Olivares-Gazca, J.C.; Priesca-Marín, J.M.; Ojeda-Laguna, M.; Garces-Eisele, J.; Soto-Olvera, S.; Palacios-Alonso, A.; Izquierdo-Vega, J.; Chacon-Cano, R.; Arizpe-Bravo, D.; López-Trujillo, M.A. Infusion of convalescent plasma is associated with clinical improvement in critically ill patients with COVID-19: A pilot study. Rev. Investig. Clin. 2020, 72, 159–164. [Google Scholar] [CrossRef]
  83. Tremblay, D.; Seah, C.; Schneider, T.; Bhalla, S.; Feld, J.; Naymagon, L.; Wang, B.; Patel, V.; Jun, T.; Jandl, T. Convalescent Plasma for the Treatment of Severe COVID-19 Infection in Cancer Patients. Cancer Med. 2020, 9, 8571–8578. [Google Scholar] [CrossRef] [PubMed]
  84. Ye, M.; Fu, D.; Ren, Y.; Wang, F.; Wang, D.; Zhang, F.; Xia, X.; Lv, T. Treatment with convalescent plasma for COVID-19 patients in Wuhan, China. J. Med. Virol. 2020, 92, 1890–1901. [Google Scholar] [CrossRef] [Green Version]
  85. Pham, H.P.; Staley, E.M.; Raju, D.; Marin, M.J.; Kim, C.H. Laboratory Assay Evaluation Demystified: A Review of Key Factors Influencing Interpretation of Test Results Using Different Assays for SARS-CoV-2 Infection Diagnosis. Lab. Med. 2020, 51, e66–e70. [Google Scholar] [CrossRef]
  86. Føns, S.; Krogfelt, K.A. How can we interpret SARS-CoV-2 antibody test results? Pathog. Dis. 2021, 79, ftaa069. [Google Scholar] [CrossRef]
  87. Brochot, E.; Demey, B.; Handala, L.; François, C.; Duverlie, G.; Castelain, S. Comparison of different serological assays for SARS-CoV-2 in real life. J. Clin. Virol. 2020, 130, 104569. [Google Scholar] [CrossRef]
  88. Menis, M.; Sridhar, G.; Selvam, N.; Ovanesov, M.V.; Divan, H.A.; Liang, Y.; Scott, D.; Golding, B.; Forshee, R.; Ball, R. Hyperimmune globulins and same-day thrombotic adverse events as recorded in a large healthcare database during 2008–2011. Am. J. Hematol. 2013, 88, 1035–1040. [Google Scholar] [CrossRef] [PubMed]
  89. Dzik, S. COVID-19 convalescent plasma: Now is the time for better science. Transfus. Med. Rev. 2020, 34, 141–144. [Google Scholar] [CrossRef]
  90. Takada, A.; Feldmann, H.; Ksiazek, T.G.; Kawaoka, Y. Antibody-dependent enhancement of Ebola virus infection. J. Virol. 2003, 77, 7539–7544. [Google Scholar] [CrossRef] [Green Version]
  91. Nakajima, K. Donor complications and donor care. ISBT Sci. Ser. 2009, 4, 411–417. [Google Scholar] [CrossRef]
  92. Davey, R.T., Jr.; Fernández-Cruz, E.; Markowitz, N.; Pett, S.; Babiker, A.G.; Wentworth, D.; Khurana, S.; Engen, N.; Gordin, F.; Jain, M.K. Anti-influenza hyperimmune intravenous immunoglobulin for adults with influenza A or B infection (FLU-IVIG): A double-blind, randomised, placebo-controlled trial. Lancet Respir. Med. 2019, 7, 951–963. [Google Scholar] [CrossRef] [Green Version]
  93. Hung, I.F.; To, K.K.; Lee, C.-K.; Lee, K.-L.; Yan, W.-W.; Chan, K.; Chan, W.-M.; Ngai, C.-W.; Law, K.-I.; Chow, F.-L. Hyperimmune IV immunoglobulin treatment: A multicenter double-blind randomized controlled trial for patients with severe 2009 influenza A (H1N1) infection. Chest 2013, 144, 464–473. [Google Scholar] [CrossRef] [PubMed]
  94. Rodriguez, W.J.; Gruber, W.C.; Groothuis, J.R.; Simoes, E.A.; Rosas, A.J.; Lepow, M.; Hemming, V.; Group, R.-I.S. Respiratory syncytial virus immune globulin treatment of RSV lower respiratory tract infection in previously healthy children. Pediatrics 1997, 100, 937–942. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Boxplots of mortality rates in COVID-19 patients from control and CPT recipient groups of RCTs, MCSs, and CRs. The total number of patients included in each category of studies is labeled above as N = 319, 9655, and 170 for the RCTs, MCSs, and CRs, respectively.
Figure 1. Boxplots of mortality rates in COVID-19 patients from control and CPT recipient groups of RCTs, MCSs, and CRs. The total number of patients included in each category of studies is labeled above as N = 319, 9655, and 170 for the RCTs, MCSs, and CRs, respectively.
Viruses 13 00849 g001
Table 1. Studies showing effects of convalescent plasma therapy in COVID-19 cases.
Table 1. Studies showing effects of convalescent plasma therapy in COVID-19 cases.
Study, CountryCPT (and Control)
Patient Count
CPT and (Control) Mortality
(%)
CPT and (Control) Cohort Age GroupAntibody Threshold aPlasma
Volume (mL)
Follow-Up Days
Randomized Clinical Trials (RCTs)
[49], Spain38; (43)0; (9)61; (60)-250–30029
[51], Netherlands43; (43)14; (26)63; (61)NA(t) ≥ 1:80≥30030
[26], China52; (51)16; (24)70; (69)NA(t) ≥ 1:64020028
[50], Iraq21; (28)5; (29)56; (48)EIgG(i) > 1.25≥20030
Matched-Control Studies (MCSs)
[61], Iran115; (74)15; (24)54; (57)EIgG(i) > 1.1≥50030
[63], Turkey888; (888)25; (28)60; (61)-200–60017
[56], USA47; (1340)23; (42)59; (-)NA(t) ≥ 1:500≥20030
[42], China10; (10)0; (30)53; (53)NA(t) ≥ 1:640200-
[54], USA20; (20)10; (30)60; (-)-≥20014
[64], USA35,322; (-) b[8.3–26.7] c; (-)60; (-)S/Co [4.62–18.45]2007
[59], USA39; (156)13; (24)55; (54)EIgG(t) ≥ 1:32050014
[53], Qatar40; (40)3; (13)48; (56)-40028
[52], Italy46; (23)7; (30)63; (-)NA(t) ≥ 1:80≥2507
[62], USA64; (177)13; (16)61; (61)-≥20028
[57], Argentina868; (2298)25; (44)56; (64)EIgG(i) > 1350200–25028
[58], USA321; (582)6; (12)53; (60)EIgG(i) ≥ 1350≥20060
[60], China138; (1430)2; (4)65; (63)-≥20014
[55], China6; (15)83; (93) d62; (73)-≥200-
Case Reports (CRs)
[70], South Korea2069-25015
[75], USA1035-40010
[76], China1038-30031
[71], Hungary20--60014
[65], China16065TA [10.9–115 AU/mL]200–12008
[77], Turkey1055EIgG(i) > 1.135011
[80], USA3113--≥2007
[79], France17658NA(t) ≥ 1:408007
[72], South Korea1068-50023
[81], USA291758-20028
[67], China6061NA(t) ≥ 1:4020060
[68], USA3024-40031
[78], China10100NA(t) ≥ 1:64030013
[66], Mexico8057EIgG(t) > 1:10050023
[82], Mexico102052-≥2008
[73], China1066EIgG(t) > 1:16040026
[43], China5065NA(t) ≥ 1:4040047
[83], USA244269EIgG(t) ≥ 1:3205009
[74], China1065-80011
[84], China6058-≥20025
[69], China4057-≥20038
a Different serologic tests to report antibody levels across studies, including the enzyme-linked immunosorbent assay for immunoglobulin G (EIgG) and neutralizing antibody (NA) indices (i) or titers (t), signal-to-cutoff ratio (S/Co), and total antibodies (TA) in arbitrary units per volume (AU/mL). b Joyner et al. [64] reports on an Expanded Access Program for the treatment of COVID-19 patients with CPT. The program did not include a control group and was, therefore, excluded from the boxplot and random-effects model. c Range of 7- and 30-day mortality rates corresponding to different times of transfusion (<3 or ≥4 days) after diagnosis and antibody S/Co levels. d The CPT and control group mortality rates (83 and 93%) reported by Zeng et al. [55] are not shown as outliers in Figure 1 for visualization purposes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kanj, S.; Al-Omari, B. Convalescent Plasma Transfusion for the Treatment of COVID-19 in Adults: A Global Perspective. Viruses 2021, 13, 849. https://0-doi-org.brum.beds.ac.uk/10.3390/v13050849

AMA Style

Kanj S, Al-Omari B. Convalescent Plasma Transfusion for the Treatment of COVID-19 in Adults: A Global Perspective. Viruses. 2021; 13(5):849. https://0-doi-org.brum.beds.ac.uk/10.3390/v13050849

Chicago/Turabian Style

Kanj, Saly, and Basem Al-Omari. 2021. "Convalescent Plasma Transfusion for the Treatment of COVID-19 in Adults: A Global Perspective" Viruses 13, no. 5: 849. https://0-doi-org.brum.beds.ac.uk/10.3390/v13050849

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop