Next Article in Journal
Artificial Intelligence Model to Detect Real Contact Relationship between Mandibular Third Molars and Inferior Alveolar Nerve Based on Panoramic Radiographs
Next Article in Special Issue
Chronic Autoimmune Gastritis: Modern Diagnostic Principles
Previous Article in Journal
Mechanical Properties of Lumbar and Cervical Paravertebral Muscles in Patients with Axial Spondyloarthritis: A Case-Control Study
Previous Article in Special Issue
Comparative Evaluation of Standard E TB-Feron ELISA and QuantiFERON-TB Gold Plus Assays in Patients with Tuberculosis and Healthcare Workers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Safety and Efficacy of Convalescent Plasma in COVID-19: An Overview of Systematic Reviews

1
Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, 46100 Mantua, Italy
2
Santorso Hospital, AULSS7 Pedemontana, 36061 Vicenza, Italy
3
North-Western Tuscany Blood Bank, Pisa University Hospital, 56126 Pisa, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2021, 11(9), 1663; https://doi.org/10.3390/diagnostics11091663
Submission received: 19 August 2021 / Revised: 6 September 2021 / Accepted: 9 September 2021 / Published: 11 September 2021
(This article belongs to the Special Issue Diagnostic Infectious Disease and Microbiology)

Abstract

:
Convalescent plasma (CP) from patients recovered from COVID-19 is one of the most studied anti-viral therapies against SARS-COV-2 infection. The aim of this study is to summarize the evidence from the available systematic reviews on the efficacy and safety of CP in COVID-19 through an overview of the published systematic reviews (SRs). A systematic literature search was conducted up to August 2021 in Embase, PubMed, Web of Science, Cochrane and Medrxiv databases to identify systematic reviews focusing on CP use in COVID-19. Two review authors independently evaluated reviews for inclusion, extracted data and assessed quality of evidence using AMSTAR (A Measurement Tool to Assess Reviews) and GRADE tools. The following outcomes were analyzed: mortality, viral clearance, clinical improvement, length of hospital stay, adverse reactions. In addition, where possible, subgroup analyses were performed according to study design (e.g., RCTs vs. non-RCTs), CP neutralizing antibody titer and timing of administration, and disease severity. The methodological quality of included studies was assessed using the checklist for systematic reviews AMSTAR-2 and the GRADE assessment. Overall, 29 SRs met the inclusion criteria based on 53 unique primary studies (17 RCT and 36 non-RCT). Limitations to the methodological quality of reviews most commonly related to absence of a protocol (11/29) and funding sources of primary studies (27/29). Of the 89 analyses on which GRADE judgements were made, effect estimates were judged to be of high/moderate certainty in four analyses, moderate in 38, low in 38, very low in nine. Despite the variability in the certainty of the evidence, mostly related to the risk of bias and inconsistency, the results of this umbrella review highlight a mortality reduction in CP over standard therapy when administered early and at high titer, without increased adverse reactions.

1. Introduction

The Coronavirus Disease 2019 (COVID-19) pandemic, caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is still a worldwide health crisis with devastating social and economic consequences. Despite the virus known for more than a year and a half, the management of COVID-19 remains challenging with still high mortality rates among severely affected patients [1,2]. A number of guidelines from experts have been released during the pandemic period suggesting several treatments for COVID-19 patients, including antiviral, hydroxychloroquine, steroid, anticoagulation and other supportive therapies [3,4]. However, recent evidence from large-scale studies failed to clarify the efficacy of most of the treatments proposed [5,6,7].
Convalescent plasma (CP), first introduced in 1890 by Emil von Behring to treat diphtheria and pertussis and then utilized in several other serious infectious diseases, including Ebola, severe acute respiratory syndrome (SARS), and Middle East respiratory syndrome (MERS), has been also proposed more recently as passive immunotherapy for treatment of SARS-CoV-2 infection [1]. Convalescent plasma is nowadays among the most studied and utilized antibody-based therapies against COVID-19 world-wide and a number of published or ongoing clinical trials have been conducted to assess its efficacy and safety in this challenging viral infection. Their conclusions are, however, quite conflicting and reflect the wide heterogeneity between different studies in terms of CP product, patients enrolled, and disease characteristics. Because of the huge amount of clinical data available, several systematic reviews (SRs) and meta-analysis have been published in the last year to harmonize the results from primary clinical trials. To synthesize the evidence from these SRs and meta-analyses, we have decided to apply to this clinical setting a relatively new approach, i.e., to perform an overview of the existing SRs, also called umbrella review.

2. Material and Methods

This umbrella review was registered at the International Prospective Register of Systematic Reviews (PROSPERO) with the registration number CRD42021259625.

2.1. Review Question/Objective

The aim of this umbrella review is to evaluate the efficacy and safety of CP for the treatment of COVID-19 patients.

2.2. Inclusion and Exclusion Criteria

We considered for inclusion SRs that included randomized controlled trials (RCTs) and non-RCTs (i.e., prospective, retrospective, cross-sectional, cohort studies and case series) evaluating the safety and efficacy of CP in COVID-19 patients. Reviews without qualitative and/or quantitative analysis were excluded from this umbrella review. SRs evaluating other viral infections were excluded unless they also contained data on SARS-COV-2 infection that could be evaluated separately.

2.3. Clinical Setting and Participants

For this umbrella review, we considered SRs on COVID-19 at any stage of disease severity, from asymptomatic/paucisymptomatic to life-threatening cases, and in any setting (outpatients and hospitalized patients). In addition, we included populations of patients with no limitations of age, gender, ethnicity, or comorbidities.

2.4. Intervention and Outcomes

CP treatment at any titer, dose, timing and frequency was compared to standard of care or placebo. We included the following outcomes: all-cause mortality, viral clearance, clinical improvement, length of hospital stay, serious and non-serious adverse reactions. Subgroup analyses were also performed based on the severity (i.e., non-severe versus severe) of COVID-19 patients treated with CP and on the titer (i.e., high versus low titer) and timing (i.e., <3 days versus >3 days of hospital admission) of CP transfusion.

2.5. Search Strategy

Relevant studies in four bibliographic databases (Embase, PubMed, Web of Science, and Cochrane) and a preprint database MedRix were searched as of 15 August 2021. The searches were carried-out without languages restriction using Medical Subjects Heading: (“COVID-19” OR “SARS-CoV-2”) AND (“convalescent plasma” OR “serotherapy” OR “hyperimmune plasma therapy” OR “convalescent plasma treatment”) AND (“systematic review” OR “meta-analysis”). In addition to the electronic search, we checked the reference lists of the most relevant items (original studies and reviews) to identify potentially eligible studies not captured by the initial literature search.

2.6. Study Selection and Data Extraction

All titles were screened by two independent assessors (MC and MF). Eligibility assessment was based on the title or abstract and on the full text if required. Full texts of possibly eligible articles were obtained and assessed independently by two reviewers (MC and MF). Both reviewers compared the articles identified. Studies were selected independently by two reviewers (MF and MC) with disagreements resolved through discussion and on the basis of the opinion of a third reviewer (FC). The two assessors also independently extracted quantitative and qualitative data from each selected study. Findings are presented in tabular format with supporting text (Table 1). Quantitative tabulation of results include: first author name and year of publication, the clinical condition under evaluation, principal characteristics of the study population, number of RCTs and non-RCTs included in the SR, intervention (CP versus control characteristics), the outcomes assessed, a quantitative synthesis (when available) of the estimates of interest (odds ratio (OR), risk ratio (RR), risk difference (RD), or mean difference (MD) with the 95% confidence intervals (CI), as reported in individual reviews), and the main results and conclusions of the SR. In addition, a three-color score was used for an immediate visual inspection of the CP-related effects with regard to the four main outcomes assessed: viral clearance, clinical improvement, mortality reduction and safety (green color: CP confers advantage over standard therapy or placebo; red color: CP does not confer advantage over standard therapy or placebo; yellow color: no clear advantage or disadvantage).

2.7. Assessment of Methodological Quality

We used A Measurement Tool to Assess Reviews (AMSTAR-2) critical appraisal checklist for SRs, a tool that evaluates both quantitative and qualitative reviews [8]. The tool is suitable for reviews including randomised and non-randomised studies. It includes 16 domains relating to the research question, review design, search strategy, study selection, data extraction, justification for excluded studies, description of included studies, risk of bias, sources of funding, meta-analysis, heterogeneity, publication bias, and conflicts of interest. Two review authors (MC, MF) independently assessed the quality of evidence in the included reviews and the methodological quality of the SRs. We resolved discrepancies through discussion or, if needed, through a third review author (FC). We did not exclude reviews based on AMSTAR-2 ratings, but considered the ratings in interpretation of our results.

2.8. Appraisal of the Quality of Evidence

The quality of evidence was appraised following the Grades of Recommendation, Assessment, Development, and Evaluation (GRADE) approach. Whenever available, the grading of the quality of evidence reported in the included reviews was considered to determine the quality of evidence. In a situation in which the grading of evidence was not reported by the authors of the study, the GRADE approach was applied in its five domains (risk of bias, indirectness, imprecision, inconsistency, and publication bias) based on the information available from the study [9].
Table 1. Summary of the published systematic reviews and meta-analyses.
Table 1. Summary of the published systematic reviews and meta-analyses.
First
Author
Clinical
Setting
PopulationStudies Included in Quantitative Analysis (n)InterventionOutcomesMain Results
Overall
(Patients)
RCTNRCTCPControl
Aviani [10]MERS-CoV, SARS-CoV-1, SARS-CoV-2 and influenza infections (severe or critically ill patients)Patients infected with beta coronaviruses or influenza viruses with no limitations of age, gender or ethnicity 20515CP at any titer, dose, timing and frequencyStandard care30-day mortality, safety, viral clearance, clinical improvement, discharge rateCP significantly reduced mortality and increased the number of discharged patients. Less than 1% of serious transfusion-related AEs
Bansal [11]COVID-19 at any stage of disease severityAdult (>18 years) patients hospitalized for COVID-1923
(27,706)
1013CP at any titer, dose, timing and frequencyPlacebo or standard therapyMortality, safetyCP significantly reduced mortality. A 6.1% pooled AE rate was observed with no CP-related fatalities
Barreira [12]Moderate, severe or critical COVID-19Hospitalized COVID-19 patients with no limitations of age, gender or ethnicity11
(3098)
56CP at any titer, dose, timing and frequencyPlacebo or standard careMortality, safety, viral clearance, clinical improvement, length of hospitalizationCP significantly reduced mortality and viral load at 72 h after CP transfusion. A 3.5% AE rate occurred
De Candia [13]COVID-19 at any stage of disease severityPatients with laboratory confirmed COVID-1925
(22,591)
1015CP at any titer, dose, timing and frequencyStandard therapyMortalityCP, independently from neutralizing titer, significantly reduced mortality only when administered at early stage of the disease
Elbadawi [14]COVID-19 pneumoniaCOVID-19 patients 6
(1226)
60CP at any titer, dose, timing and frequencyStandard careAll-cause mortality, progression to severe respiratory illness, clinical improvement, need for IMV, safetyCP did not reduce all-cause mortality, the progression to sever respiratory illness or the need for IMV. No differences in clinical improvement and AEs
Gupta [15]COVID-19 at any stage of disease severityCOVID-19 patients12
(13,206)
120CP at any titer, dose, timing and frequencyPlacebo or standard care28-day mortality, clinical improvement, viral clearance, safetyCP was not associatd with clinical improvement or significantly reduced risk of death. A low incidence (3.2%) of AEs was observed
Janiaud [16]COVID-19 at any stage of disease severityPatients with confirmed or suspected COVID-19 in any treatment setting10
(11,782)
100CP at any titer, dose, timing and frequencyPlacebo + standard therapy or standard therapyClinical improvement, all-cause mortality, no. of patients requiring IMV, rate of serious adverse eventsCP did not reduce the need for IMV and all-cause mortality. A subgroup analysis showed a mortality reduction in patients receiving high-titer CP [17]
Juul [18]COVID-19 at any stage of disease severityCOVID-19 patients with no limitations of age, gender or ethnicity33
(13,312)
33
(2 CP)
0Various interventions for treatment of COVID-19, including CP, were analyzedStandard careAll-cause mortality, admission to ICU, need for IMV, non-serious AEsCP did not reduce all-cause mortality
Keikha [19] MERS-CoV (2 studies), SARS-CoV-1 (5 studies), SARS-CoV-2 (8 studies) infectionsPatients infected with beta coronaviruses15
(5240)
114CP at any titer, dose, timing and frequencyPlacebo or standard therapyClinical improvement, viral clearance,
hospital discharge mortality
The clinical improvement was significantly increased in CP group versus control group
Kim [20]Moderate-severe COVID-19Adult (> 18 years) patients hospitalized for COVID-19110
(4 CP)
40
(2 CP)
70
(2 CP)
Various pharmacological interventions against COVID-19, including CP, were analyzedPlacebo or standard careMortality, progression to severe disease, viral clearance, serious adverse eventsCP was associated with significantly reduced mortality rate in non-ICU setting and improved viral clearance rate at 2 weeks compared to standard care
Klassen [21]COVID-19 at any stage of disease severityCOVID-19 patients with no limitations of age, gender or ethnicity301020CP at any titer, dose and timing (<3 days versus >3 days of hospital admission)Standard careMortalityCP significantly reduced mortality rate compared with standard care. CP transfusion within 3 days of hospital admission resulted in greater mortality reduction. A subgroup analysis documented the safety of CP [22]
Kloypan [23]COVID-19 at any stage of disease severityCOVID-19 patients471433CP at any titer, dose, timing and frequencyPlacebo or standard treatment28-day mortality, length of hospital stay, clinical improvement, discharge rateCP significantly reduced the risk of all-cause mortality compared to standard care
Meher [24]Moderate-severe COVID-19Hospitalized adult patients of both gender with moderate to
severe COVID-19
6
(474)
24CP at any titer, dose, timing and frequencyStandard careMortality, clinical improvement, viral clearanceCP significantly reduced all-cause mortality and viral detection by day 7 with no clinical improvement by day 7
Peng [25]Prophylaxis and treatment of COVID-19COVID-19 patients with no limitations of age, gender, ethnicity or underlying diseases13
(2984)
211CP at any titer, dose, timing and frequencyPlacebo or standard careMortality, clinical improvement, safetyCP significanlty reduced mortality and increased viral clearance
Piechotta [26]COVID-19 at any stage of disease severity (asymptomatic
or symptomatic)
COVID-19 patients with no limitations of age, gender or ethnicity13
(48,509)
121CP at any titer, dose, timing and frequencyPlacebo or standard careAll-cause mortality, clinical improvement, need for IMV, viral clearance, safetyCP did not reduce the need for IMV and 28-day all-cause mortality, but increased 7-day viral clearance. In a subgroup analysis, CP decreased disease progression and all-cause mortality in individuals with asymptomatic or mild COVID-19
Prasad [27]Severe and non-severe COVID-19Hospitalized COVID-19 patients with no limitations of age, gender or ethnicity22
(9622)
913CP at any titer, dose, timing and frequencyPlacebo or standard careMortality, clinical improvement, need for IMV, viral clearance,
length of ICU or hospital stay, safety
Inconclusive effects of CP on mortality, clinical improvement, need for mechanical ventilation and faster viral clearance
Rabelo-da-Ponte [28]COVID-19 at any stage of disease severityCOVID-19 patients with no limitations of age, gender or ethnicity9
(149)
18CP at any titer, dose, timing and frequencyStandard treatmentViral clearance, clinical improvementCP reduced viral load and was associated with clinical status improvement
Sarkar [29]COVID-19 at any stage of disease severityCOVID-19 patients with no limitations of age, gender or ethnicity7
(5444)
25CP at any titer, dose, timing and frequencyStandard treatmentMortality, viral clearance, clinical improvementCP reduced mortality, increased viral clearance and was associated with clinical improvement
Sun [30]Different types of infectious diseases including severe COVID-19Patients with viral infections with no limitations of age and sex153
(1 CP)
12CP at any titer, dose, timing and frequencyStandard treatmentMortality, symptom duration, hospital length of stay, antibody levels, viral load, adverse eventsThere was a significantly lower mortality rate in the group treated with CP compared with control groups
Talaie [31]COVID-19 at any stage of disease severityAdult COVID-19 patients26
(3263)
14
(1 CP)
12
(5 CP)
Various pharmacological interventions against COVID-19, including CP, were analyzedStandard treatmentMortality, viral clearance, clinical improvement, ICU entry, need for IMVCP had a beneficial effect on clinical improvement and negative seroconversion and tended to decrease mortality
Vegivinti [32]COVID-19 at any stage of disease severityCOVID-19 patients with no limitations of age, gender or ethnicity15
(4898)
510CP at any titer, dose, timing and frequencyStandard careMortality, clinical improvement,
length of hospital stay
CP was associated with a significantly reduced mortality and higher clinical improvement
Wang [33]COVID-19 at any stage of disease severityAdult (>18 years) COVID-19 patients42
(8 CP)
10
(1 CP)
32
(7 CP)
CP at any titer, dose, timing and frequencyStandard careMortality,
viral clearance
CP tended to decrease the mortality risk and was associated with a higher viral nucleic acid negative rate
Wang [34]COVID-19 at any stage of disease severityCOVID-19 patients with no limitations of age, gender or ethnicity45
(44,068)
441CP at any titer, dose, timing and frequencyPlacebo, standard care, no interventionMortality,
clinical improvement, safety
CP reduced (NRCTs) or not (RCTs) mortality and improved (NRCTs) or not (RCTs) clinical symptoms
Wardhani [35]Mild and severe COVID-19 COVID-19 patients12
(5342)
39CP at any titer, dose, timing and frequencyStandard careAll-cause mortality, subgroup analysis based on disease severityAll and severe COVID-19 patients not receiving CP were at increased mortality risk compared to those treated with CP
Wenjing [36]Severe and critical COVID-19Severely and critically ill COVID-19 patients with no limitations of age, gender or ethnicity936CP at any titer, dose, timing and frequencyNAMortality, clinical improvement,
safety
CP significantly reduced mortality. A qualitative analysis showed a beneficial effect of CP in reducing viral load, on clinical improvement and on safety
Yuwono Soeroto [37]COVID-19 at any stage of disease severityCOVID-19 patients18
(5658)
711CP at any titer, dose, timing and frequencyStandard careMortalityCP use was associated with significantly decreased mortality
Zhang [38]Severe COVID-19Critically ill COVID-19 patients with no limitations of age, gender or ethnicity19118CP at any titer, dose, timing and frequencyPlacebo or standard careMortality, safety, viral clearanceA significantly reduced mortality rate and a higher negative rate of PCR was found in the CP versus control group
Abbreviations: RCT, randomized controlled trial; NRCT, non-randomized controlled trial; CP, convalescent plasma; NA, not available; IMV, invasive mechanical ventilation; Severe acute respiratory syndrome coronavirus: SARS; Middle East respiratory syndrome: MERS; ICU, intensive care unit; PCR, polymerase chain reaction; AE, adverse event.

3. Results

The electronic and manual search retrieved 244 references. At the first stage of screening titles and abstracts, 52 references were selected. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow diagram is reported in Figure 1. After the full texts were scrutinized against the inclusion and exclusion criteria, 29 SRs were included in the umbrella review [10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38] and 23 SRs were excluded [39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61]. Reasons for exclusion were: SRs not covering or with no informative data on CP therapy in COVID-19 [39,40,41,42,44,45,47,48,49,54] and reviews on CP therapy in COVID-19 with no quantitative and/or qualitative analysis [43,46,50,51,52,53,55,56,57,58,59,60,61].

3.1. Description of the Studies

Of the 29 SRs included in the overview, 26 were focused exclusively on COVID-19 [11,12,13,14,15,16,17,18,20,21,22,23,24,25,26,27,28,29,31,32,33,34,35,36,37,38], while three were focused on respiratory pandemics and on beta coronaviruses infections [10,19,30]. Two SRs [17,22] were a subgroup analysis of other reviews [16,21]. The 29 SRs included 653 overlapping reports (237 RCTs and 416 non-RCTs), based on 53 individual primary studies. The primary studies included 17 RCTs, 26 controlled non-RCTs, and 10 uncontrolled studies (single arm studies, including case series and case reports). Twenty-eight SRs were focused on CP treatment of COVID-19, while one study [25] was focused on both CP prophylaxis and treatment. The majority of the SRs analyzed COVID-19 at any stage of disease severity, while four studies [10,30,36,38] were focused on more advanced (severe and/or critical) stages. Most SRs analyzed COVID-19 patients with no limitation of age, gender or ethnicity, while four SRs were focused only on adult (>18 years) COVID-19 patients [11,20,31,33]. The main characteristics of the SRs included are summarized in Table 1.

3.2. Methodological Quality

Of the included reviews, the majority (19/29; 65.5%) had ≥2 (from 2 to 12) unmet AMSTAR-2 methodological requirements, and nine (31.0%) had one unmet methodological requirement; one Cochrane review met all the methodologic requirements (Table 2). Sixteen reviews (55.2%) had one or more methodological requirements partly met. Twenty-seven reviews (93.1%) did not report on the source of funding for the studies included in the review; 11 reviews (39.2%) did not register or publish a protocol. Eight reviews (27.6%) did not mention publication bias in methods and results, and failed to discuss the possible impact of publication bias on review findings. In five reviews, participants, interventions, comparators, and outcomes (PICO) were not clearly made explicit, and in 3 reviews design was not fully explained. In six reviews, the search strategy was not comprehensive. More than 85% of author teams perform study selection and screening in duplicate. Other unmet domains were related to the list of excluded reviews and reasons (seven reviews, 24.1%), assessment of risk of bias (three reviews), and sources of conflict of interest, including any funding that the authors receive for conducting the review (four reviews).

3.3. Summary of the Effect of CP on the Main Outcomes

3.3.1. (a) Outcome “Overall Mortality”

Overall mortality was the most common reported outcome. Great heterogeneity was ascertained in several SRs; thus, we performed subgroup analyses to control for sources of heterogeneity such as design of studies included in the review (e.g., RCTs and non-RCTs), titre of SARS-CoV-2 neutralizing antibodies (high or low), and time of administration (early or late). The results of our analyses are summarized in Table 3. Sixteen SRs reported the outcome mortality in RCTs and non-RCT. In 14 of these SRs the effect size favoured the CP arm compared to controls, while in two SRs it was unclear whether CP reduced mortality compared to controls; the quality of the evidence was very low in 2 SRs, low in four, moderate in nine, and from moderate to high in one. Eleven SRs analysed the outcome mortality in RCTs only, and all were consistent in concluding that CP did not reduce mortality compared to controls (moderate certainty of evidence in 10 SRs, from moderate to high certainty in one SR). The analysis of results from non-RCTs showed higher reduction in mortality in CP group compared to control in six out of seven SRs (low quality of evidence in four, very low in one, moderate in two).
Due to the clinical heterogeneity observed in many of the primary studies and statistical heterogeneity in many of the SRs, we performed subgroup analyses as specified in the protocol. High titre CP was more effective than lower titre in reducing mortality (moderate certainty of evidence in three SRs, low in one). In two SRs, it was unclear whether high titre CP reduces mortality compared to controls (low certainty of evidence). Subgroup analysis according to time to CP transfusion showed a reduction in mortality in early CP recipients compared to controls in three SRs (moderate certainty of evidence); by contrast a Cochrane review based on a single RCT (RECOVERY trial) [62] concluded that it is unclear whether early CP transfusion reduces mortality compared to controls (low quality of evidence), although transfusion after the first week of illness resulted in higher risk of mortality compared to early transfusion. We also performed subgroup analysis of mortality according to baseline severity of COVID-19, but there was heterogeneity in defining the clinical condition. In patients with moderate/severe infection, the effect of CP was unclear (three SRs, low-quality of evidence), while in patients with severe/critical infection, the results were more heterogeneous in the comparison, effect size and certainty of the evidence (Table 3).

3.3.2. (b) Outcome “Viral Clearance”

The outcome viral clearance (rate of patients with negative reverse transcription polymerase chain reaction (RT-PCR) test for SARS-CoV-2 after a positive test at baseline) was reported in 6 SRs after 3 days, and in two SRs after 7–14 days (Table 4). On day 3, four out of six SRs reported an increase in viral clearance in CP recipients compared to controls (from low to moderate certainty of evidence), while in two SRs it was unclear whether CP reduced the viral clearance compared to controls (from low to moderate certainty of evidence). On day 14, CP showed significantly higher viral clearance rate compared to standard supportive therapy in one SR (low certainty of evidence), while in a Cochrane review based on two RCTs it was unclear whether CP increases viral clearance compared to controls (low certainty of evidence). It was not possible to perform subgroup analyses for these outcomes due to the relatively low number of primary studies and SRs available.

3.3.3. (c) Outcome “Clinical Improvement”

Clinical improvement was reported in nine SRs. Six SRs concluded that it is unclear whether CP increases rate of clinical improvement compared to controls (in three SRs moderate certainty of evidence, in three low certainty of evidence). Three SRs showed that CP increase rate of clinical improvement compared to controls (low certainty of evidence in two SRs, moderate certainty of evidence in one SR) (see Table 4).

3.3.4. (d) Outcome “Length of Hospital Stay”

Length of hospital stay was reported in six SRs. All concluded that it is unclear whether CP decreases length of hospital stay compared to controls (low quality of certainty in four SRs, moderate certainty of evidence in two SRs) (Table 4).

3.3.5. (e) Outcome “Adverse Events”

Serious adverse events to CP transfusion were reported in five SRs and overall adverse events in two SRs. All SRs concluded that the frequency of adverse reactions was similar in CP and control groups (low quality of certainty in four SRs, moderate certainty of evidence in one SR) (Table 5).

3.4. GRADE Assessment

Of the 89 analyses on which GRADE judgements were made, effect estimates were judged to be of high/moderate certainty in four analyses, moderate in 38, low in 38, and very low in nine. For the outcome mortality, the judgment was very low in nine analyses, low in 19, moderate in 19 and moderate/high in four.

4. Discussion

The outbreak of the COVID-19 pandemic has greatly accelerated the clinical trial research evaluating the safety and efficacy of CP as emergency therapy. According to the study by Muller-Olling and colleagues [63], CP is the second most frequent investigational medicinal product evaluated in COVID-19-related clinical trials and increasing interest in this form of immunotherapy is documented by the fact that more than 140 clinical trials specifically evaluating CP in COVID-19 have been registered to date worldwide [63]. Studies on CP as treatment for COVID-19 have qualitatively evolved during the COVID-19 pandemic period in response to the advances in the knowledge of this disease and to the results of the published clinical trials [64]. They can be generally classified into three generations: the first-generation clinical trials, performed at the beginning of the first pandemic wave, utilized CP with high-titer anti-SARS-CoV-2 neutralizing antibodies usually in hospitalized patients with severe, advanced COVID-19 [65,66]. However, it soon became evident that, since CP was working by blocking the viral replication, the earlier it was used the more effective it was in preventing the disease progression [1]. Thus, second generation clinical trials were focused on the use of high-titer CP early in the course of COVID-19 (within 3 days from symptom onset or hospitalization) [67]. Finally, to optimize the possible beneficial effect of plasma, the more recent third generation trials are evaluating the CP infusion in particular populations of patients at high risk of development of severe or critical COVID-19, such as those with impaired humoral immunity, onco-hematological disorders or other severe cardiovascular or respiratory co-morbidities [68,69,70]. Moreover, some of these trials encompass the pre-transfusion evaluation of recipients’ anti-SARS-CoV-2 antibody levels in order to capture those patients with lack or insufficient antibody response and therefore most likely to benefit from CP passive immunotherapy [1]. Based on the above, it is evident that the clinical trials conducted during the 18-month COVID-19 pandemic are widely heterogeneous in terms of study design and CP administration schedule, disease and patients characteristics. In this extremely uncertain and changing context, typical of emergency situations such as those of the COVID-19 pandemic, it is evident that even the systematic reviews and meta-analyses have produced heterogeneous results. This overview of reviews includes data from twenty-nine systematic reviews, based on more than 600 overlapping reports and 53 individual primary studies (43 controlled trials, including 17 RCTs and 26 non-RCTs, and 10 uncontrolled trials (single arm studies). We believe that makes this the largest review to date within this subject area, and hope this will make it particularly helpful to decision makers. The results of RCTs are not always consistent with the results of observational studies, and differences in estimated magnitude of treatment effect are very common, often resulting in overestimation of treatment effects in observational studies [71]. Interpretation of the results obtained from both RCTs and observational studies, as well as from systematic reviews including both types of study design, can help understand the efficacy/effectiveness and safety of a therapeutic option [72]. For this reason we performed, where available, subgroup analyses of the effect size obtained in the overall comparison, in RCTs and in observational studies. For the outcome most commonly reported, overall mortality, it was possible to perform subgroup analysis of SRs according to study design, antibodies titre and time of transfusion. While the majority of SRs reporting this outcome in non-RCTs and in non-RCTs + RCTs showed a reduction in mortality in CP recipients compared to controls, when the analysis was limited to RCTs it was unclear whether CP reduced mortality compared to controls. It was also clear that most of the included studies (both RCTs and non-RCTs) were at risk of bias and showed significant clinical, methodological and statistical heterogeneity. Overall the certainty of the evidence was from low to moderate in the majority of the SRs. Subgroup analyses according to neutralizing antibody titres and time of CP transfusion showed a reduction in mortality in the majority of SRs when high titres of antibody and early transfusions were administered (from low to moderate certainty of the evidence). The other secondary outcomes (i.e., viral clearance, clinical improvement and length of hospital stay) were addressed by only a minority of SRs with a high level of uncertainty, so that no definitive conclusions can be drawn. However, CP seemed to be effective in increasing viral clearance as compared with standard therapy, particularly within the first three days from CP transfusion (from low to moderate certainty of the evidence). Additionally, CP did not increase the risk of adverse events between intervention and control groups, confirming the safety of this procedure [22]. Although the SRs tried to address, at least in part, the heterogeneity of the results (on the basis of neutralizing titre, time of CP infusion and study design) it was almost impossible to evaluate in this overview the clinical heterogeneity of primary studies, related to different disease conditions at baseline, concomitant therapies, patients characteristics. Limitations to the methodological quality of reviews most commonly related to absence of a protocol (11/29) and funding sources of primary studies (27/29).
In conclusion, despite these limitations and based on the analysis of the main outcome mortality, this overview of systematic reviews supports the safety and efficacy of the clinical use of CP over standard therapy when administered at high titer and early during the course of COVID-19. Further pooled qualitative and quantitative analyses from a new systematic review based on individual patients data rather than on aggregate data (that are often insufficient for a thorough analysis) or from adequately powered third generation clinical trials (i.e., assessing the early use of high titer CP administered in populations of patients with inadequate antiviral response and at increased risk of developing severe COVID-19) are needed to pinpoint exactly where and when CP can give the greatest clinical benefit in COVID-19.

Author Contributions

Conceptualization: M.C and M.F. Methodology: M.C. Validation: M.F. and F.C. Writing—preparation original draft: M.C. and M.F. Writing—review and editing: F.C. and D.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest in regard to this work.

References

  1. Franchini, M.; Liumbruno, G.; Piacentini, G.; Glingani, C.; Zaffanello, M. The Three Pillars of COVID-19 Convalescent Plasma Therapy. Life 2021, 11, 354. [Google Scholar] [CrossRef] [PubMed]
  2. Baud, D.; Qi, X.; Nielsen-Saines, K.; Musso, D.; Pomar, L.; Favre, G. Real estimates of mortality following COVID-19 infection. Lancet Infect. Dis. 2020, 20, 773. [Google Scholar] [CrossRef] [Green Version]
  3. Falavigna, M.; Colpani, V.; Stein, C.; Azevedo, L.C.P.; Bagattini, A.M.; De Brito, G.V.; Chatkin, J.M.; Cimerman, S.; Corradi, M.D.F.D.B.; Da Cunha, C.A.; et al. Guidelines for the pharmacological treatment of COVID-19. The task force/consensus guideline of the Brazilian Association of Intensive Care Medicine, the Brazilian Society of Infectious Diseases and the Brazilian Society of Pulmonology and Tisiology. Rev. Bras. Ter. Intensiv. 2020, 32, 166–196. [Google Scholar] [CrossRef]
  4. Rochwerg, B.; Siemieniuk, R.A.; Agoritsas, T.; Lamontagne, F.; Askie, L.; Lytvyn, L.; Agarwal, A.; Leo, Y.-S.; Macdonald, H.; Zeng, L.; et al. A living WHO guideline on drugs for covid-19. BMJ 2020, 370, m3379. [Google Scholar] [CrossRef]
  5. Singh, A.K.; Singh, A.; Singh, R.; Misra, A. Hydroxychloroquine in patients with COVID-19: A Systematic Review and meta-analysis. Diabetes Metab. Syndr. 2020, 14, 589–596. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, Y.; Zhang, D.; Du, G.; Du, R.; Zhao, J.; Jin, Y.; Fu, S.; Gao, L.; Cheng, Z.; Lu, Q.; et al. Remdesivir in adults with severe COVID-19: A randomised, double-blind, placebo-controlled, multicentre trial. Lancet 2020, 395, 1569–1578. [Google Scholar] [CrossRef]
  7. WHO Solidarity Trial Consortium. Repurposed Antiviral Drugs for Covid-19-Interim WHO Solidarity Trial Results. N. Engl. J. Med. 2021, 384, 497–511. [Google Scholar] [CrossRef] [PubMed]
  8. Shea, B.J.; Reeves, B.C.; Wells, G.; Thuku, M.; Hamel, C.; Moran, J.; Moher, D.; Tugwell, P.; Welch, V.; Kristjansson, E.; et al. AMSTAR 2: A critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. BMJ 2017, 358, j4008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Schünemann, H.; Brożek, J.; Guyatt, G.; Oxman, A. (Eds.) Handbook for Grading the Quality of Evidence and the Strength of Recommendations Using the GRADE Approach; Updated October 2013; GRADE Working Group: Hamilton, ON, Canada, 2013; Available online: https://gradepro.org/handbook/ (accessed on 17 August 2021).
  10. Aviani, J.K.; Halim, D.; Soeroto, A.Y.; Achmad, T.H.; Djuwantono, T. Current views on the potentials of convalescent plasma therapy (CPT) as Coronavirus disease 2019 (COVID-19) treatment: A systematic review and meta-analysis based on recent studies and previous respiratory pandemics. Rev. Med. Virol. 2021. [Google Scholar] [CrossRef]
  11. Bansal, V.; Mahapure, K.S.; Mehra, I.; Bhurwal, A.; Tekin, A.; Singh, R.; Gupta, I.; Rathore, S.S.; Khan, H.; Deshpande, S.; et al. Mortality Benefit of Convalescent Plasma in COVID-19: A Systematic Review and Meta-Analysis. Front. Med. 2021, 8, 624924. [Google Scholar] [CrossRef]
  12. Barreira, D.F.; Lourenço, R.A.; Calisto, R.; Moreira-Gonçalves, D.; Santos, L.L.; Videira, P.A. Assessment of the Safety and Therapeutic Benefits of Convalescent Plasma in COVID-19 Treatment: A Systematic Review and Meta-Analysis. Front. Med. 2021, 8, 660688. [Google Scholar] [CrossRef] [PubMed]
  13. de Candia, P.; Prattichizzo, F.; Garavelli, S.; La Grotta, R.; De Rosa, A.; Pontarelli, A.; Parrella, R.; Ceriello, A.; Matarese, G. Effect of time and titer in convalescent plasma therapy for COVID-19. iScience 2021, 24, 102898. [Google Scholar] [CrossRef] [PubMed]
  14. Elbadawi, A.; Shnoda, M.; Laguio-Vila, M.; Mamas, M.A.; Elgendy, I.Y. Convalescent plasma in the management of COVID-19 pneumonia. Eur. J. Intern. Med. 2021, 89, 121–123. [Google Scholar] [CrossRef] [PubMed]
  15. Gupta, T.; Kannan, S.; Kalra, B.; Thakkar, P. Systematic review and meta-analysis of randomised controlled trials testing the safety and efficacy of convalescent plasma in the treatment of coronavirus disease 2019 (COVID-19): Evidence-base for practise and implications for research. Transfus. Med. 2021. [Google Scholar] [CrossRef]
  16. Janiaud, P.; Axfors, C.; Schmitt, A.M.; Gloy, V.; Ebrahimi, F.; Hepprich, M.; Smith, E.R.; Haber, N.A.; Khanna, N.; Moher, D.; et al. Association of Convalescent Plasma Treatment with Clinical Outcomes in Patients with COVID-19: A Systematic Review and Meta-Analysis. JAMA 2021, 325, 1185–1195. [Google Scholar] [CrossRef] [PubMed]
  17. Cruciani, M.; Bongiovanni, G.; Franchini, M. High-titer convalescent plasma therapy for coronavirus disease 2019 and mortality. Transfusion 2021, 61, 1988–1990. [Google Scholar] [CrossRef]
  18. Juul, S.; Nielsen, E.E.; Feinberg, J.; Siddiqui, F.; Jørgensen, C.K.; Barot, E.; Nielsen, N.; Bentzer, P.; Veroniki, A.A.; Thabane, L.; et al. Interventions for treatment of COVID-19: A living systematic review with meta-analyses and trial sequential analyses (The LIVING Project). PLoS Med. 2020, 17, e1003293. [Google Scholar] [CrossRef]
  19. Keikha, M.; Karbalaei, M. Convalescent plasma therapy as a conventional trick for treating COVID-19: A systematic review and meta-analysis study. New Microbes New Infect. 2021, 42, 100901. [Google Scholar] [CrossRef]
  20. Kim, M.S.; An, M.H.; Kim, W.J.; Hwang, T.-H. Comparative efficacy and safety of pharmacological interventions for the treatment of COVID-19: A systematic review and network meta-analysis. PLoS Med. 2020, 17, e1003501. [Google Scholar] [CrossRef]
  21. Klassen, S.A.; Senefeld, J.W.; Johnson, P.W.; Carter, R.E.; Wiggins, C.C.; Shoham, S.; Grossman, B.J.; Henderson, J.P.; Musser, J.; Salazar, E.; et al. The Effect of Convalescent Plasma Therapy on Mortality Among Patients With COVID-19: Systematic Review and Meta-analysis. Mayo Clin. Proc. 2021, 96, 1262–1275. [Google Scholar] [CrossRef]
  22. Franchini, M.; Cruciani, M. How safe is COVID-19 convalescent plasma? Mayo Clin. Proc. 2021, 96, 2279–2281. [Google Scholar] [CrossRef]
  23. Kloypan, C.; Saesong, M.; Sangsuemoon, J.; Chantharit, P.; Mongkhon, P. CONVALESCENT plasma for COVID-19: A meta-analysis of clinical trials and real-world evidence. Eur. J. Clin. Investig. 2021, e13663. [Google Scholar] [CrossRef]
  24. Meher, B.R.; Padhy, B.M.; Das, S.; Mohanty, R.R.; Agrawal, K. Effectiveness of Convalescent Plasma Therapy in the Treatment of Moderate to Severe COVID 19 Patients: A Systematic Review and Meta-Analysis. J. Assoc. Phys. India 2020, 68, 35–43. [Google Scholar]
  25. Peng, H.T.; Rhind, S.G.; Beckett, A. Convalescent Plasma for the Prevention and Treatment of COVID-19: A Systematic Review and Quantitative Analysis. JMIR Public Health Surveill. 2021, 7, e25500. [Google Scholar] [CrossRef] [PubMed]
  26. Piechotta, V.; Iannizzi, C.; Chai, K.L.; Valk, S.J.; Kimber, C.; Dorando, E.; Monsef, I.; Wood, E.M.; Lamikanra, A.A.; Roberts, D.J.; et al. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: A living systematic review. Cochrane Database Syst Rev. 2021, 5, CD013600. [Google Scholar]
  27. Prasad, M.; Seth, T.; Elavarasi, A. Efficacy and Safety of Convalescent Plasma for COVID-19: A Systematic Review and Meta-analysis. Indian J. Hematol. Blood Transfus. 2021, 37, 347–365. [Google Scholar] [CrossRef] [PubMed]
  28. Rabelo-Da-Ponte, F.D.; Silvello, D.; Scherer, J.N.; Ayala, A.R.; Klamt, F. Convalescent Plasma Therapy in Patients with Severe or Life-Threatening COVID-19: A Metadata Analysis. J. Infect. Dis. 2020, 222, 1575–1578. [Google Scholar] [CrossRef]
  29. Sarkar, S.; Soni, K.D.; Khanna, P. Convalescent plasma is a clutch at straws in COVID-19 management! A systematic review and meta-analysis. J. Med. Virol. 2020, 93, 1111–1118. [Google Scholar] [CrossRef]
  30. Sun, M.; Xu, Y.; He, H.; Zhang, L.; Wang, X.; Qiu, Q.; Sun, C.; Guo, Y.; Qiu, S.; Ma, K. A potentially effective treatment for COVID-19: A systematic review and meta-analysis of convalescent plasma therapy in treating severe infectious disease. Int. J. Infect. Dis. 2020, 98, 334–346. [Google Scholar] [CrossRef] [PubMed]
  31. Talaie, H.; Hosseini, S.M.; Nazari, M.; Fakhri, Y.; Mousavizadeh, A.; Vatanpour, H.; Firoozfar, A. Is there any potential management against COVID-19? A systematic review and meta-analysis. DARU J. Pharm. Sci. 2020, 28, 765–777. [Google Scholar] [CrossRef]
  32. Vegivinti, C.T.R.; Pederson, J.M.; Saravu, K.; Gupta, N.; Evanson, K.W.; Kamrowski, S.; Schmidt, M.; Barrett, A.; Trent, H.; Dibas, M.; et al. Efficacy of convalescent plasma therapy for COVID-19: A systematic review and meta-analysis. J. Clin. Apher. 2021, 36, 470–482. [Google Scholar] [CrossRef]
  33. Wang, M.; Wu, T.; Zuo, Z.; You, Y.; Yang, X.; Pan, L.; Hu, Y.; Luo, X.; Jiang, L.; Xia, Z.; et al. Evaluation of current medical approaches for COVID-19: A systematic review and meta-analysis. BMJ Support. Palliat. Care 2020, 11, 45–52. [Google Scholar] [CrossRef]
  34. 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. 2020, 91, 107262. [Google Scholar] [CrossRef]
  35. Wardhani, S.O.; Fajar, J.K.; Wulandari, L.; Soegiarto, G.; Purnamasari, Y.; Asmiragani, A.; Maliga, H.A.; Ilmawan, M.; Seran, G.; Iskandar, D.S.; et al. Association between convalescent plasma and the risk of mortality among patients with COVID-19: A meta-analysis. F1000Research 2021, 10, 64. [Google Scholar] [CrossRef] [PubMed]
  36. Wenjing, L.; Yuanzheng, F.; Li, J.-Y.; Tang, L.V.; Yu, H. Safety and efficacy of convalescent plasma therapy in severely and critically ill patients with COVID-19: A systematic review with meta-analysis. Aging 2020, 13, 1498–1509. [Google Scholar] [CrossRef] [PubMed]
  37. Soeroto, A.Y.; Purwiga, A.; Alam, A.; Prasetya, D. Plasma convalescent decrease mortality in COVID-19 patients: A systematic review and meta-analysis. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 4841–4853. [Google Scholar]
  38. Zhang, X.; Xi, L.; Pang, F.; Du, Y.; Yuan, Q.; Shi, M.; Liu, J.; Ma, H.; Li, B. Convalescent Plasma in the Treatment of Severe COVID-19: A Systematic Review and Meta-Analysis. Iran. J. Public Health 2020, 49, 2022–2031. [Google Scholar] [CrossRef] [PubMed]
  39. Abeldaño Zuñiga, R.A.; Coca, S.M.; Abeldaño, G.F.; González-Villoria, R.A.M. Clinical effectiveness of drugs in hospitalized patients with COVID-19: A systematic review and meta-analysis. Ther. Adv. Respir. Dis. 2021, 15, 17534666211007214. [Google Scholar]
  40. Abubakar, A.R.; Sani, I.H.; Godman, B.; Kumar, S.; Islam, S.; Jahan, I.; Haque, M. Systematic Review on the Therapeutic Options for COVID-19: Clinical Evidence of Drug Efficacy and Implications. Infect. Drug Resist. 2020, 13, 4673–4695. [Google Scholar] [CrossRef] [PubMed]
  41. Alunno, A.; Najm, A.; Mariette, X.; De Marco, G.; Emmel, J.; Mason, L.; McGonagle, D.G.; Machado, P.M. Immunomodulatory therapies for SARS-CoV-2 infection: A systematic literature review to inform EULAR points to consider. Ann. Rheum. Dis. 2021, 80, 803–815. [Google Scholar] [CrossRef] [PubMed]
  42. AminJafari, A.; Ghasemi, S. The possible of immunotherapy for COVID-19: A systematic review. Int. Immunopharmacol. 2020, 83, 106455. [Google Scholar] [CrossRef]
  43. Bakhtawar, N.; Usman, M.; Khan, M.M.U. Convalescent Plasma Therapy and Its Effects On COVID-19 Patient Outcomes: A Systematic Review of Current Literature. Cureus 2020, 12, e9535. [Google Scholar] [CrossRef] [PubMed]
  44. Ray, S.; Haldar, R.; Choupoo, N.S.; Das, S.K.; Sarkar, H.; Tewari, R. Evaluating the Efficacy and Safety of the Existing Repurposed Pharmacological Agents for Treating COVID-19: A Meta-analysis and Systematic Review of Clinical Trials. Indian J. Crit. Care Med. 2020, 24, 1106–1113. [Google Scholar] [CrossRef]
  45. Devasenapathy, N.; Ye, Z.; Loeb, M.; Fang, F.; Najafabadi, B.T.; Xiao, Y.; Couban, R.; Bégin, P.; Guyatt, G. Efficacy and safety of convalescent plasma for severe COVID-19 based on evidence in other severe respiratory viral infections: A systematic review and meta-analysis. CMAJ 2020, 192, E745–E755. [Google Scholar] [CrossRef]
  46. Fabricius, M.M.; Dandachi, D. COVID-19 Convalescent Plasma: From donation to treatment–A Systematic Review & Single Center Experience. Mo. Med. 2021, 118, 74–80. [Google Scholar] [PubMed]
  47. Han, Y.J.; Lee, K.H.; Yoon, S.; Nam, S.W.; Ryu, S.; Seong, D.; Kim, J.S.; Lee, J.Y.; Yang, J.W.; Lee, J.; et al. Treatment of severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and coronavirus disease 2019 (COVID-19): A systematic review of In Vitro, In Vivo, and clinical trials. Theranostics 2021, 11, 1207–1231. [Google Scholar] [CrossRef] [PubMed]
  48. Heustess, A.; Allard, M.; Thompson, D.; Fasinu, P. Clinical Management of COVID-19: A Review of Pharmacological Treatment Options. Pharmaceuticals 2021, 14, 520. [Google Scholar] [CrossRef]
  49. Khani, E.; Khiali, S.; Entezari-Maleki, T. Potential COVID-19 Therapeutic Agents and Vaccines: An Evidence-Based Review. J. Clin. Pharmacol. 2021, 61, 429–460. [Google Scholar] [CrossRef]
  50. Mansourabadi, A.H.; Sadeghalvad, M.; Mohammadi-Motlagh, H.R.; Rezaei, N. The immune system as a target for therapy of SARS-CoV-2: A systematic review of the current immunotherapies for COVID-19. Life Sci. 2020, 258, 118185. [Google Scholar] [CrossRef]
  51. Meo, S.A.; Zaidi, S.Z.A.; Shang, T.; Zhang, J.Y.; Al-Khlaiwi, T.; Bukhari, I.A.; Akram, J.; Klonoff, D.C. Biological, molecular and pharmacological characteristics of chloroquine, hydroxychloroquine, convalescent plasma, and remdesivir for COVID-19 pandemic: A comparative analysis. J. King Saud Univ. Sci. 2020, 32, 3159–3166. [Google Scholar] [CrossRef]
  52. Munir, M.; Tandiabang, P.; Setyawati, T.; Basry, A.; Cyio, A.; Rahman, N. Bioethical perspective of convalescent plasma therapy for COVID-19: A systematic review. Transfus. Clin. Biol. 2021, 28, 271–275. [Google Scholar] [CrossRef]
  53. Nassar, M.; Nso, N.; Alfishawy, M.; Novikov, A.; Yaghi, S.; Medina, L.; Toz, B.; Lakhdar, S.; Idrees, Z.; Kim, Y.; et al. Current systematic reviews and meta-analyses of COVID-19. World J. Virol. 2021, 10, 182–208. [Google Scholar] [CrossRef] [PubMed]
  54. Raja, M.A.; Mendoza, M.A.; Villavicencio, A.; Anjan, S.; Reynolds, J.M.; Kittipibul, V.; Fernandez, A.; Guerra, G.; Camargo, J.F.; Simkins, J.; et al. COVID-19 in solid organ transplant recipients: A systematic review and meta-analysis of current literature. Transplant. Rev. 2020, 35, 100588. [Google Scholar] [CrossRef] [PubMed]
  55. 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] [PubMed]
  56. Seth, T.; Elavarasi, A.; Sahoo, R.K.; Madan, K.S.; Nischal, N.; Soneja, M.; Garg, P.; Prasad, K.; Sharma, A. Convalescent plasma therapy for Covid-19: A systematic review. Natl. Med. J. India. 2020, 33, 213–221. [Google Scholar]
  57. Shao, S.; Wang, Y.; Kang, H.; Tong, Z. Effect of convalescent blood products for patients with severe acute respiratory infections of viral etiology: A systematic review and meta-analysis. Int. J. Infect. Dis. 2020, 102, 397–411. [Google Scholar] [CrossRef]
  58. Chandrasekar, V.T.; Venkatesalu, B.; Patel, H.K.; Spadaccini, M.; Manteuffel, J.; Ramesh, M. Systematic review and meta-analysis of effectiveness of treatment options against SARS-CoV-2 infection. J. Med. Virol. 2021, 93, 775–785. [Google Scholar] [CrossRef]
  59. Wadaa-Allah, A.; Emhamed, M.S.; Sadeq, M.A.; Dahman, N.B.H.; Ullah, I.; Farrag, N.S.; Negida, A. Efficacy of the current investigational drugs for the treatment of COVID-19: A scoping review. Ann. Med. 2021, 53, 318–334. [Google Scholar] [CrossRef]
  60. Wooding, D.J.; Bach, H. Treatment of COVID-19 with convalescent plasma: Lessons from past coronavirus outbreaks. Clin. Microbiol. Infect. 2020, 26, 1436–1446. [Google Scholar] [CrossRef]
  61. Zaffanello, M.; Piacentini, G.; Nosetti, L.; Franchini, M. The use of convalescent plasma for pediatric patients with SARS-CoV-2: A systematic literature review. Transfus. Apher. Sci. 2021, 60, 103043. [Google Scholar] [CrossRef]
  62. RECOVERY Collaborative Group. Convalescent plasma in patients admitted to hospital with COVID-19 (RECOVERY): A randomised controlled, open-label, platform trial. Lancet 2021, 397, 2049–2059. [Google Scholar] [CrossRef]
  63. Müller-Olling, M.; Vahlensieck, U.; Hilger, A. Heterogeneity in COVID-19 Convalescent Plasma Clinical Trials. Clin. Pharmacol. Ther. 2021. [Google Scholar] [CrossRef]
  64. Casadevall, A.; Grossman, B.J.; Henderson, J.P.; Joyner, M.J.; Shoham, S.; Pirofski, L.-A.; Paneth, N. The Assessment of Convalescent Plasma Efficacy against COVID-19. Med 2020, 1, 66–77. [Google Scholar] [CrossRef]
  65. 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, 324, 460–470. [Google Scholar] [CrossRef] [PubMed]
  66. Simonovich, V.A.; Pratx, L.D.B.; Scibona, P.; Beruto, M.V.; Vallone, M.G.; Vázquez, C.; Savoy, N.; Giunta, D.H.; Pérez, L.G.; Sánchez, M.D.L.; et al. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N. Engl. J. Med. 2021, 384, 619–629. [Google Scholar] [CrossRef]
  67. Libster, R.; Marc, G.P.; Wappner, D.; Coviello, S.; Bianchi, A.; Braem, V.; Esteban, I.; Caballero, M.T.; Wood, C.; Berrueta, M.; et al. Early High-Titer Plasma Therapy to Prevent Severe Covid-19 in Older Adults. N. Engl. J. Med. 2021, 384, 610–618. [Google Scholar] [CrossRef] [PubMed]
  68. Thompson, M.A.; Henderson, J.P.; Shah, P.K.; Rubinstein, S.M.; Joyner, M.J.; Choueiri, T.K.; Flora, D.B.; Griffiths, E.A.; Gulati, A.P.; Hwang, C.; et al. Association of Convalescent Plasma Therapy with Survival in Patients with Hematologic Cancers and COVID-19. JAMA Oncol. 2021, 7, 1167–1175. [Google Scholar] [CrossRef] [PubMed]
  69. Focosi, D.; Franchini, M. Potential use of convalescent plasma for SARS-CoV-2 prophylaxis and treatment in immunocompromised and vulnerable populations. Expert Rev. Vaccines 2021, 1–8. [Google Scholar] [CrossRef]
  70. Cohn, C.S.; Estcourt, L.; Grossman, B.J.; Pagano, M.B.; Allen, E.S.; Bloch, E.M.; Casadevall, A.; Devine, D.V.; Dunbar, N.M.; Foroutan, F.; et al. COVID -19 convalescent plasma: Interim recommendations from the AABB. Transfusion 2021, 61, 1313–1323. [Google Scholar] [CrossRef]
  71. Ioannidis, J.P.A.; Haidich, A.-B.; Pappa, M.; Pantazis, N.; Kokoris, S.; Tektonidou, M.; Contopoulos-Ioannidis, D.G.; Lau, J. Comparison of Evidence of Treatment Effects in Randomized and Nonrandomized Studies. JAMA 2001, 286, 821–830. [Google Scholar] [CrossRef]
  72. Faraoni, D.; Schaefer, S.T. Randomized controlled trials vs. observational studies: Why not just live together? BMC Anesthesiol. 2016, 16, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. PRISMA flow diagram of study selection.
Figure 1. PRISMA flow diagram of study selection.
Diagnostics 11 01663 g001
Table 2. The AMSTAR-2 checklist.
Table 2. The AMSTAR-2 checklist.
Author [Reference]AMSTAR-2 DOMAIN
12345678910111213141516
Aviani [10]
Bansal [11]
Barreira [12]
De Candia [13]
Elbadawi [14]
Gupta [15]
Janiaud [16]
Cruciani [17]
Juul [18]
Keikha [19]
Kim [20]
Klassen [21]
Franchini [22]
Kloypan [23]
Meher [24]
Peng [25]
Piechotta [26]
Prasad [27]
Rabelo-da-Ponte [28]
Sarkar [29]
Sun [30]
Talaie [31]
Vegivinti [32]
Wang M [33]
Wang Y [34]
Wardhani [35]
Wenjing [36]
Yuwono Soeroto [37]
Zhang [38]
Footnotes: Diagnostics 11 01663 i001 Methodological requirement met, Diagnostics 11 01663 i002 Methodological requirement partly met, Diagnostics 11 01663 i003 Methodological requirement unmet. AMSTAR-2 domains: 1. Did the research questions and inclusion criteria for the review include the components of PICO? 2. Did the report of the review contain an explicit statement that the review methods were established prior to the conduct of the review and did the report justify any significant deviations from the protocol? 3. Did the review authors explain their selection of the study designs for inclusion in the review? 4. Did the review authors use a comprehensive literature search strategy? 5. Did the review authors perform study selection in duplicate? 6. Did the review authors perform data extraction in duplicate? 7. Did the review authors provide a list of excluded studies and justify the exclusions? 8. Did the review authors describe the included studies in adequate detail? 9. Did the review authors use a satisfactory technique for assessing the risk of bias (RoB) in individual studies that were included in the review? 10. Did the review authors report on the sources of funding for the studies included in the review? 11. If meta-analysis was performed did the review authors use appropriate methods for statistical combination of results? 12. If meta-analysis was performed, did the review authors assess the potential impact of RoB in individual studies on the results of the meta-analysis or other evidence synthesis? 13. Did the review authors account for RoB in individual studies when interpreting/ discussing the results of the review? 14. Did the review authors provide a satisfactory explanation for, and discussion of, any heterogeneity 15. If they performed quantitative synthesis did the review authors carry out an adequate investigation of publication bias (small study bias) and discuss its likely impact on the results of the review? 16. Did the review authors report any potential sources of conflict of interest, including any funding they received for conducting the review? Although AMSTAR-2 consists of 16 items, critical domains include items 2, 4, 7, 9, 11, 13, and 15.
Table 3. Effects of convalescent plasma on overall mortality.
Table 3. Effects of convalescent plasma on overall mortality.
Review
[Reference]
No. Studies No. Subjects
(CP/Controls)
Effect Size (RR, OR or RD) and 95% CIs GRADE
Assessment
CommentEffect
Direction
Outcome: MortalityOverall Analysis (RCTs + non-RCTs)
Aviani [10]12 (4/8)4306 (724/3582)RR 0.62 (0.46/0.82)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Bansal [11]23 (11/12)7542 (2392/5150)OR 0.65 (0.53/0.80)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Barreira [12]11 (5/6)2998 (823/2175)RR 0.71 (0.57/0.90)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
De Candia [13]25 (10/15)24772 (13470/11302)RR 0.78 (0.68/0.90)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Klassen [21]30 (10/20)12982 (1425/11467)OR 0.58 (0.47/0.71)From moderate
⊕⊕⊕⊝
to high ⊕⊕⊕⊕ 3
CP reduces mortality compared to controls
Kloypan [23]20 (11/9)16533 (7753/8780)RR 0.69 (0.56/0.86)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Meher [24]6 (2/4)469 (167/302)RR 0.61 (0.37/0.99)⊕⊝⊝⊝
Very low 4
CP reduces mortality compared to controls
Peng [25]13 (4/9)2984 (695/2289)OR 0.48 (0.34/0.67)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Sarkar [29]7 (2/5)5454 (5169/285)OR 0.44 (0.25/0.77)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Talaie [31]3 (1/2)163 (82/81)RR 0.52 (0.26/1.03)⊕⊝⊝⊝
Very low 5
It is unclear whether CP reduces mortality compared to controls
Vegivinti [32]15 (5/10)4858 (2208/2650)OR 0.58 (0.44/0.78)⊕⊝⊝⊝
Very low 4
CP reduces mortality compared to controls
Wang Y [34]3 (1/2)319 (97/222)RR 0.65 (0.42/1.02)⊕⊝⊝⊝
Very low 5
It is unclear whether CP reduces mortality compared to controls
Wardhani [35]12 (3/9)5342 (1937/3405)OR 1.92 (1.33/2.77)⊕⊕⊝⊝
Low 1
Mortality higher in controls
Wenjing [36]10 (3/7)2835 (2271/564)RR 0.57 (0.44/0.74)⊕⊝⊝⊝
Very low 4
CP reduces mortality compared to controls
Yuwono Soeroto [37]18 (6/12)5657 (2168/3489)OR 0.64 (0.49/0.84)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Zhang [38]4 (1/3)182 (87/95)RR 0.59 (0.37/0.94)⊕⊝⊝⊝
Very low 5
CP reduces mortality compared to controls
RCTs only
Bansal [11]101413 (770/653)OR 0.75 (0.53/1.08)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP reduces mortality compared to controls
Barreira [12]51065 (595/470)OR 0.85 (0.62/1.16)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP reduces mortality compared to controls
De Candia [13]1013470 (6579/6891)RR 0.96 (0.91/1.03)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP reduces mortality compared to controls
Elbadawi [14]61307 (737/550)OR 0.83 (0.58/1.18)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP reduces mortality compared to controls
Gupta [15]1213204 (6715/6489)RR 0.81 (0.65/1.02)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP reduces mortality compared to controls
Janiaud [16]91384 (758/626)RR 1.02 (0.92/1.12)⊕⊕⊕⊝
Moderate 6
It is unclear whether CP reduces mortality compared to controls
Klassen [21]101425 (771/654)OR 0.76 (0.54/1.09)From moderate ⊕⊕⊕⊝
to high ⊕⊕⊕⊕ 3
It is unclear whether CP reduces mortality compared to controls
Meher [24]2187 (94/93)RR 0.60 (0.33/1.10)⊕⊕⊕⊝
Moderate 6
It is unclear whether CP reduces mortality compared to controls
Prasad [27]81336 (730/606)OR 0.85 (0.61/1.18)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP reduces mortality compared to controls
Wang M [33]2167 (81/86)OR 0.40 (0.14/1.11)⊕⊕⊕⊝
Moderate 6
It is unclear whether CP reduces mortality compared to controls
Yuwono Soeroto [37]61142 ()647/495)RR 0.85 (0.71/1.02)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP reduces mortality compared to controls
Non-RCTs only
Bansal [11]94087 (1278/2809)OR 0.78 (0.65/0.94)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Barreira [12]62033 (328/1705)RR 0.56 (0.39/0.81)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Klassen [21]2011467 (3150/8317)OR 0.57 (0.45/0.72)From moderate ⊕⊕⊕⊝
To high ⊕⊕⊕⊕ 3
CP reduces mortality compared to controls
Meher [24]4273 (73/200)OR 0.48 (0.17/1.36)⊕⊝⊝⊝
Very low 4
It is unclear whether CP reduces mortality compared to controls
Prasad [27]138267 (2621/5646)OR 0.66 (0.53/0.82)⊕⊕⊝⊝
Low 1
CP reduces mortality compared to controls
Wang M [33]117779 (1649/6130)RR 0.59 (0.53/0.66)⊕⊝⊝⊝
Very low 5
CP reduces mortality compared to controls
Yuwono Soeroto [37]124515 (1521/2994)RR 0.48 (0.34/0.70)⊕⊕⊕⊝
Moderate 2
CP reduces mortality compared to controls
Subgroup analysis: High antibody titre
Aviani [10]High titre (>640)
-3 studies (2 RCTs) in severely ill pts
-2 studies (1 RCT) in critical ill pts
1186 (98/1088)388 (43/345)RR 0.42 (0.22/0.78)RR 0.72 (0.46/1.12)⊕⊕⊕⊝
Moderate 2
⊕⊕⊝⊝
Low 7
-In pts with severe illness, high titre CP reduces mortality compared to controls. The reduction in mortality in study with lower antibody titre (neutralizing titre ≤ 1:320) was less marked (RR 0.80, 95% CI 0.47/1.34).
-In pts with critical illness, it is unclear whether CP reduces mortality compared to controls
Barreira [12]High titre (>1:297) (4 studies, 2 RCTs)650 (329/321)RR 0.68 (0.44/1.04)⊕⊕⊝⊝
Low 7
It is unclear whether high titre CP reduces mortality compared to controls. In studies with lower titre (<1:297) CP the RR was higher: 0.85 (0.58/1.25)
Cruciani [17]High titre, 3 RCTs374 (170/174)RD −0.06 (−0.12/0.00)⊕⊕⊕⊝
Moderate 2
High titre CP reduces mortality compared to controls
De Candia [13]High titre (different cut-off) (14 studies, 7 RCTs)20744 (11711/9033)RR 0.93 (0.88/0.99)⊕⊕⊕⊝
Moderate 2
High titre CP reduces mortality compared to controls
Klassen [21]2 non-RCTs1125 (534 high titre, 591 lower titre)22% high titre CP, 29% lower titre⊕⊕⊝⊝
Low 7
Mortality higher in those receiving lower CP titre transfusion
Subgroup analysis: early CP transfusion
Barreira [12]3 studies (2 RCTs)2118 (416/1702)RR 0.71 (0.53/0.96)⊕⊕⊕⊝
Moderate 2
Mortality was reduced in pts receiving early (within 7 days) CP transfusion compared to controls. In pts receiving late (> 7 days) CP transfusion mortality was similar to that observed in control group (RR 0.60, 95% CI 0.30/1.17)
De Candia [13]11 studies (5 RCTs)19007 (8018/10989)OR 0.72 (0.68/0.77)⊕⊕⊕⊝
Moderate 2
Mortality was reduced in pts receiving early (within 3 days) CP transfusion compared to controls. In pts receiving late (> 7 days) CP transfusion mortality was similar to that observed in control group (OR 0.94, 95% CI 0.86/1.04)
Klassen [21]8 studies (3 RCTs)1561 (656/905)OR 0.44 (0.32/0.61)⊕⊕⊕⊝
Moderate 2
Mortality reduction associated with convalescent plasma transfusion was greater in studies that transfused patients within 3 days of hospital admission (OR, 0.44; 95% CI 0.32–0.61) compared with studies that transfused patients more than 3 days after hospital admission (OR, 0.79; 95% CI 0.62/0.98)
Piechotta [26]1 RCT (Recovery trial)4466RR 0.93 (0.84/1.02)⊕⊕⊕⊝
Moderate 2
It is unclear whether early CP transfusion (within 7 days of symptoms onset) reduces mortality compared to controls. Transfusion of CP after 7 days of symptoms onset resulted in a RR of 1.04 (0.95/1.15)
Subgroup analysis according
to severity
of infection
-Moderate COVID-19
Barreira [12]3 (2 RCTs)545 (275/272)RR 0.96 (0.62/1.48)⊕⊕⊝⊝
Low 1
It is unclear whether CP transfusion reduces mortality in pts with moderate illness
Kim [20]3 non-RCTsNot availableOR 0.67 (0.16/2.74)⊕⊕⊝⊝
Low 8
It is unclear whether CP transfusion reduces mortality in pts with moderate illness
Piechotta (from moderate to severe) [26]1 RCT77 (36/41)RR 0.98 (0.68/1.41)⊕⊕⊝⊝
Low 9
It is unclear whether CP transfusion reduces mortality in pts with moderate/severe illness
Yuwono Soeroto (from moderate to severe) [37]6 (3 RCTs)938 (430/508)RR 0.51 (0.26/1.02)⊕⊕⊝⊝
Low 8
It is unclear whether CP transfusion reduces mortality in pts with moderate/severe illness
-Severe/
critical COVID-19
Aviani (Severe vs. critical) [10]4 (1 RCT)166 (80/86)RR 4.64 (2.12/10.0)⊕⊕⊝⊝
Low 7
In pts receiving CP, mortality higher in critical ill pts compared to severely illness
Barreira [12]4 (2 RCTs)1889 (240/1649)RR 0.84 (0.54/1.32)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP transfusion reduces mortality in pts with severe/critical illness
Wardhani [35]9 trials (3 RCTs)4164 (1458/2706)OR 1.32 (1.09/1.60)⊕⊕⊕⊝
Moderate 2
In pts with severe disease, mortality higher in controls compared to CP recipients
Wenjing [36]Severe: 4 trials (1 RCT)Critical: 3 trials (1 RCT)1420 (168/1252)171 (78/93)RR 0.54 (0.36/0.80)RR 0.72 (0.35/1.47)⊕⊕⊕⊝Moderate 2⊕⊝⊝⊝
very low 5
CP reduces mortality in pts with severe illness
It is unclear whether CP transfusion reduces mortality in pts with critical illness
Yuwono Soeroto [37]13 (6 RCTs)4899 (1718/3181)RR 0.68 (0.51/0.91)⊕⊕⊝⊝
Low 8
In pts with severe/critical illness, CP reduces mortality compared to controls
Footnotes: Diagnostics 11 01663 i001 Favouring CP, Diagnostics 11 01663 i002 Not clear effect of CP compared to controls. 1. Downgraded for risk of bias and heterogeneity. 2. Downgraded for risk of bias. 3. This judgment was based on the consistency of the results between RCTs and matched control studies and the corroborating evidence from dose–response studies and other uncontrolled case data. In aggregating data from all controlled studies, the meta-analyses provided precise estimates, did not demonstrate substantial heterogeneity, and demonstrated no evidence of publication bias. The inherent limitations of the included studies rendered the certainty of evidence judgment to be moderate to high. 4. Downgraded for risk of bias, heterogeneity and publication bias. 5. Downgraded for risk of bias, imprecision and heterogeneity. 6. Downgraded for imprecision. 7. Downgraded for risk of bias and imprecision. 8. Downgraded twice for risk of bias (confounding and publication bias). 9. Downgraded for imprecision and clinical heterogeneity.
Table 4. Effects of convalescent plasma on viral clearance, clinical improvement and length of hospital stay.
Table 4. Effects of convalescent plasma on viral clearance, clinical improvement and length of hospital stay.
Review
[Reference]
No. Studies No. Subjects
(CP/Controls)
Effect Size (RR, OR, HR or MD) and 95% CIs GRADE
Assessment
CommentEffect
Direction
Outcome:
viral clearance
-on day 3
Barreira [12]4 trials (3 RCTs)552 (276/276)RR 0.61 (0.38/0.98)⊕⊕⊕⊝
Moderate 1
Compared to standard treatment, CP increases rate of viral clearance after 3 days
Peng [25]3 trials (2 RCTs)128 (63/65)OR 26.21 (4.36/157.66)⊕⊕⊕⊝
Moderate 2
CP increases viral clearance compared to controls
Piechotta [26]4 RCTs552 (279/273)RR 1.73 (0.98/3.04)⊕⊕⊕⊝
Moderate 1
In patients with moderate/severe disease, it is unclear whether CP increases rate of viral clearance compared to controls
Prasad [27]2 RCTs551 (282/269)OR 3.62 (0.43/30.49)⊕⊕⊝⊝
Low 3
It is unclear whether CP increases rate of viral clearance compared to controls
Wang Y [34]2 trials (1 RCT)108 (53/55)RR 2.47 (1.70/3.57)⊕⊕⊝⊝
Low 4
CP increases viral clearance compared to controls
Zhang [38]2 trials (1 RCT)108 (53/55)RR 2.55 (1.76/3.70)⊕⊕⊝⊝
Low 4
CP increases viral clearance compared to controls
-on day 7–14
Kim [20]3 trials (1 RCT)NAOR 11.39 (3.91/33.18)⊕⊕⊝⊝
Low 5
On day 14, CP showed significantly higher viral clearance rate compared to standard supportive therapy
Piechotta [26]2 RCTs149 (79/70)RR 1.59 (0.74/3.43)⊕⊕⊝⊝
Low 3
On day 14, it is unclear whether CP increases viral clearance rate compared to standard supportive therapy
Outcome:
clinical improvement
Elbadawi [14]3 RCTs267 (130/137)OR 1.31 (0.78/2.22)⊕⊕⊝⊝
Low 5
It is unclear whether CP increases rate of clinical improvement compared to controls
Gupta [15]6 RCTs1106 (616/490)RR 1.02 (0.82/1.28)⊕⊕⊕⊝
Moderate 6
It is unclear whether CP increases rate of clinical improvement compared to controls after 7, 14 and 28 days from administration
Peng [25]4 trials (2 RCTs)404 (144/260)OR 1.54 (0.79/3.01)⊕⊕⊝⊝
Low 5
It is unclear whether CP increase rate of clinical improvement compared to controls
Piechotta [26]1 RCT77 (36/41)RR 1.10 (0.83/1.48)⊕⊕⊕⊝
Moderate 2
In patients with moderate/severe disease, it is unclear whether CP increases rate of clinical improvement (liberation from supplemental oxygen) compared to controls
Prasad [27]3 RCTs421 (322/199)OR 1.07 (0.86/1.34)⊕⊕⊝⊝
Low 5
It is unclear whether CP increases rate of clinical improvement compared to controls
Sarkar [29]7 trials (2 RCTs)5454OR 0.44 (0.25/0.77)⊕⊕⊕⊝
Moderate 6
CP increase rate of clinical improvement compared to controls
Talaie [31]3 trials (2 RCTs)Not availableRR 1.41 (1.01/1.98)⊕⊕⊝⊝
Low 5
CP increase rate of clinical improvement compared to controls
Vegivinti [32]7 trials (2 RCTs)1581OR 2.02 (1.54/2.65)⊕⊕⊝⊝
Low 5
CP increase rate of clinical improvement compared to controls
Wang M [33]2 RCTs189 (95/94)OR 1.21 (0.68/2.16)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP increase rate of clinical improvement compared to controls
Outcome:
length of hospital stay
Barreira [12]3 trials (1 RCT)2221 (488/1733)MD 1.94 (−3.69/7.58)⊕⊕⊝⊝
Low 5
It is unclear whether CP decreases length of hospital stay compared to controls
Janiaud [16]4 RCTs (2 published as preprints)603 (361/242)HR 1.07 (0.79/1.45)⊕⊕⊝⊝
Low 4
It is unclear whether CP decreases length of hospital stay compared to controls
Peng [25]6 trials (1 RCT)2101 (366/1735)MD 0.84 (−3.35/5.02)⊕⊕⊝⊝
Low 5
It is unclear whether CP decreases length of hospital stay compared to controls
Piechotta [26]5 RCTs683 (401/282)HR 1.15 (0.95/1.40)⊕⊕⊕⊝
Moderate 2
It is unclear whether CP decreases length of hospital stay compared to controls
Prasad [27]4 trials (3 RCTs)2602 (1365/1237)MD 0.12 (−1.69/1.93)⊕⊕⊕⊝
Moderate 7
It is unclear whether CP decreases length of hospital stay compared to controls
Vegivinti [32]6 trials (2 RCTs)2157MD −0.5 (−3.1/2.1)⊕⊕⊝⊝
Low 5
It is unclear whether CP decreases length of hospital stay compared to controls
Footnotes: Diagnostics 11 01663 i001 Favouring CP, Diagnostics 11 01663 i002 Not clear effect of CP compared to controls. 1. Downgraded for heterogeneity. 2. Downgraded for imprecision. 3. Downgraded for inconsistency and imprecision. 4. Downgraded for ROB and imprecision. 5. Downgraded for ROB and inconsistency. 6. Downgraded for ROB. 7. Downgraded for inconsistency.
Table 5. Adverse reactions related to convalescent plasma transfusion.
Table 5. Adverse reactions related to convalescent plasma transfusion.
Review
[Reference]
No. Studies No. Events/No. Patients in CP and ControlsEffect size (RR or RD) and 95% CIs GRADE
Assessment
CommentEffect
Direction
Outcome:
Adverse Events (AE)
-Serious AE
Gupta [15]11 RCTs200/6164 (3.2%)161/5826 (2.87)RR 1.14 (0.93/1.40)⊕⊕⊕⊝
Moderate 1
Serious AE were rare and with similar frequency in CP and controls
Janiaud [16]3 RCTs60/329 (18.2%)26/191
(13.6%)
RR 0.97 (0.36/2.63) ⊕⊕⊝⊝
Low 2
Serious AE had similar frequency in CP and controls
Juul [18]3 RCTs46/249 (18.4%)49/262 (18.7%)RR 0.93 (0.36/2.63)⊕⊕⊕⊝
Moderate 1
Serious AE had similar frequency in CP and controls
Franchini [22]9 RCTs106/853 (12.4%)52/616 (8.4%)RD 0.00 (−0.03/0.03)⊕⊕⊕⊝
Moderate 1
Serious AE had similar frequency in CP and controls
Piechotta [26]2 RCTs60/206 (29.1%)26/148 (17.5%)RR 1.73 (0.98/3.04)⊕⊕⊕⊝
Moderate 3
Serious AE were more common in CP compared to controls (29.1 vs 17.5%) but the difference was not statistically significant
-Overall AE
Franchini [22]8 RCTs1692/5848 (28.9%)1535/5471 (28.0%)RD 0.01 (−0.02/0.03)⊕⊕⊕⊝
Moderate 1
Overall AE had similar frequency in CP and controls (28%)
Piechotta [26]1 RCT153/228 (67%) 66/104 (63.4%)RR 1.06 (0.89/1.26)⊕⊕⊕⊝
Moderate 3
Overall AE had similar frequency in CP and controls (>60%). Likewise, grade 3–4 AE had similar frequency in CP recipients and controls (8.5 and 6.3%, respectively)
Footnotes: Diagnostics 11 01663 i002 Not clear effect of CP compared to controls. 1. Downgraded for ROB. 2. Downgraded for ROB and imprecision. 3. Downgraded for imprecision.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Franchini, M.; Corsini, F.; Focosi, D.; Cruciani, M. Safety and Efficacy of Convalescent Plasma in COVID-19: An Overview of Systematic Reviews. Diagnostics 2021, 11, 1663. https://doi.org/10.3390/diagnostics11091663

AMA Style

Franchini M, Corsini F, Focosi D, Cruciani M. Safety and Efficacy of Convalescent Plasma in COVID-19: An Overview of Systematic Reviews. Diagnostics. 2021; 11(9):1663. https://doi.org/10.3390/diagnostics11091663

Chicago/Turabian Style

Franchini, Massimo, Fabiana Corsini, Daniele Focosi, and Mario Cruciani. 2021. "Safety and Efficacy of Convalescent Plasma in COVID-19: An Overview of Systematic Reviews" Diagnostics 11, no. 9: 1663. https://doi.org/10.3390/diagnostics11091663

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