Skip to main content
Pathogens and Global Health logoLink to Pathogens and Global Health
. 2021 Jun 15;115(7-8):456–466. doi: 10.1080/20477724.2021.1936818

Hydroxychloroquine and mortality in COVID-19 patients: a systematic review and a meta-analysis of observational studies and randomized controlled trials

Augusto Di Castelnuovo a, Simona Costanzo b, Antonio Cassone c, Roberto Cauda d, Giovanni De Gaetano b, Licia Iacoviello b,e,
PMCID: PMC8220439  PMID: 34128772

ABSTRACT

Background: Hydroxychloroquine (HCQ) was proposed as potential treatment for COVID-19, but its association with mortality is unclear. We reviewed published literature for evidence of an association between HCQ (with or without azithromycin (AZM)) and total mortality in COVID-19 patients.

Methods: Articles were retrieved until April 29th, 2021 by searching in seven databases. Data were combined using the general-variance-based method.

Results: A total of 25 cohort studies (N=41,339 patients) and 11 randomized clinical trials (RCTs; N=8,709) were found. The use of HCQ was not associated with mortality in meta-analysis of RCTs (pooled risk ratio (PRR): 1.08, 95%CI: 0.97-1.20; I2=0%), but it was associated with 20% lower mortality risk (PRR=0.80, 95%CI: 0.69-0.93; I2=80%) in pooling of cohort studies. The negative association with mortality was mainly apparent by pooling cohort studies that used lower doses of HCQ (≤400 mg/day; PRR=0.69, 95%CI: 0.57-0.87). Use of HCQ+AZM (11 studies) was associated with 25% non-statistically significant lower mortality risk (PPR=0.75; 0.51-1.10; P=0.15). Use of HCQ was not associated with severe adverse events (PRR=1.12, 95%CI: 0.88-1.44; I2=0%).

Conclusions: HCQ use was not associated with mortality in COVID-19 patients in pooling results from RCTs (high level of certainty of evidence), but it was associated with 20% mortality reduction when findings from observational studies were combined (low level of certainty of evidence). The reduction of mortality was mainly apparent in observational studies where lower doses of HCQ were used. These findings might help disentangling the debate on HCQ use in COVID-19.

KEYWORDS: SARS-COV-2, COVID-19, hydroxychloroquine, mortality

Introduction

The aminoquinoline hydroxychloroquine (HCQ) is an anti-malaria drug with immunomodulatory and anti-thrombotic properties, currently used in the treatment of autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus and anti-phospholipid syndrome [1,2]. At the beginning of the pandemic, it was proposed as a possible therapy in COVID-19 patients since it could directly inhibit viral entry and spread in several in vitro and in vivo models [3]. Indeed, HCQ has been used in Ebola virus disease [4], human immunodeficiency virus (HIV) infection [5], SARS-CoV-1 infection and the Middle East Respiratory Syndrome [6].

Despite the lack of evidence of efficacy from few randomized clinical trials, HCQ became very popular and widely used by many clinicians. In Italy over 70% of COVID-19 hospitalized patients were treated with HCQ during the first wave of pandemic [7]. The publication of a very questionable study [8] by one of the most reliable scientific journals showing that the use of HCQ was associated to an increased risk of death, lead to the pausing of several clinical trials, including the Solidarity trial [9]. The study [8] was retracted 13 days after publication [10]. Several national agencies for drug regulation decided to suspend the authorization to use HCQ for COVID-19 treatment or prophylaxis.

Several observational studies and RCTs have been published aimed at investigating the association of HCQ use and mortality in COVID-19 patients [11]. However, a number of questions remain open on the relationship between HCQ treatments in COVID-19 patients: is there a dose issue? Does mortality rate of a population or the severity of the disease affect HCQ efficacy? Is there any interaction with other anti-COVID19 drugs? More recently, at least three large, well-conducted observational studies have been published showing that HCQ decreases mortality risk in hospitalized COVID-19 patients [7,12,13]. All these studies, which have not been included in previous meta-analysis [11], used HCQ doses lower than those administered in randomized clinical trial (RCT), such as the Solidarity or the Recovery trials [9,14].

Therefore, we decided to conduct an updated meta-analysis on observational and RCT studies on HCQ use and the mortality outcome in patients hospitalized for COVID-19. We also performed subgroup analyses to dissect whether treatment effects differ according to characteristics of the primary studies (quality of studies, peer-reviewing status, level of adjustment, sample size, setting and the effects of HCQ dosage).

Methods

This systematic review with meta-analysis was conducted according to the recommendations outlined in the Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0, and reported in line with the PRISMA statement. Institutional review board approval was not required as the study did not directly involve human participants.

Search strategy and data extraction

Flow diagram for study selection is reported in Figure 1. Articles published in English were retrieved from inception to March 17th, 2021 by searching in Medline, Embase, PubMed, Web of Science, Cochrane Central Database, MedRvix and Preprints.org, with the search terms: ‘(COVID-19 OR Cov-Sars-2) AND (hydroxychloroquine OR chloroquine)’. In addition, the reference lists of relevant articles for potential studies were also manually reviewed. After initial search, the duplicate results were removed. The remaining articles were screened for relevance by their titles and abstracts by two of us independently (SC and ADC). All selected potential articles were then reviewed by the remaining investigators to ensure their eligibility for inclusion. Disagreements about eligibility of the literature were resolved by consensus based on the agreements of all investigators.

Figure 1.

Figure 1.

Flow diagram for study selection

AZM means azithromycin; HCQ means hydroxychloroquine

To be included in this meta-analysis, the study had to meet the following criteria: (1) clinical trials or cross-sectional studies or cohort studies; (2) quantitatively investigating the difference in mortality risk in unselected COVID-19 patients according to use or not of HCQ. We only included studies in which HCQ was being used therapeutically, and excluded studies of prophylaxis.

Forty-two articles were identified [7,9,12–51]. For 11 of them [12,15,17,19,21,23,25,31,32,34,47] it was possible to extract data necessary for comparing HCQ+AZM versus no HCQ+AZM. For all other studies, it was not possible to systematically distinguish if HCQ therapy was complemented or not with AZM. Two investigators (SC and ADC) independently assessed the methodological quality of each included study by using the Joanna Briggs Institute Critical Appraisal Checklists [52], developed to assess quality of non-randomized studies such as cohort and cross-sectional studies. Each item of the checklist scored ‘0’ if it was answered ‘no’, it scored ‘1’ if it was answered ‘unclear’, if the item was answered ‘yes’, it scored ‘2’. A score higher than 80% of the total has been used as indicator of a better methodology quality and a low risk of bias [52]. Certainty of evidence was assessed using the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) approach [53]. This method evaluates the certainty of evidence by assessing the following domains: study limitations, directness, consistency, precision, and publication bias and, as additional domains, dose-response association, plausible confounding that would decrease observed effect and strength of association. Disagreements were resolved by consensus or by a third investigator (LI), if consensus could not be reached.

Data analysis

For each study, odds ratio (OR) or hazard ratio (HR) and/or number of events (number of deaths and number of total COVID-19 patients) in both the HCQ (or HCQ+AZM) and respective control groups were extracted. If available, measure of association adjusted for covariates were retrieved. Number of events were used to calculate relative risk and 95% confidence intervals (CIs) when other measures of association were not available from the primary study. The following information was also extracted: study design, if the article was not peer-reviewed, region, level of adjustment, sample size, mortality rate in the entire cohort, percentage of patients treated with HCQ, mean duration of the treatment, mean daily dose after the first day and mean total dose of HCQ used. The total dose of HCQ was calculated as the sum of the amount of drug used in the first day plus daily dose multiplied by number of days of treatment after the first.

Pre-specified subgroup analyses have been conducted for all the additional characteristics retrieved.

All analyses were performed using standard statistical procedures provided in RevMan5.1 (The Cochrane Collaboration, Oxford, United Kingdom). Data were combined using the general variance-based method, that requires information on the relative risk (or OR or HR) estimate and their 95% CI for each study. Heterogeneity was assessed using the Higgin’s I2 metric. Fixed and random effects were considered, but due to the large heterogeneity observed in cohort studies, findings from random effects were considered as primary analysis. Five studies [29,40–42,50] reported zero deaths in HCQ and/or control group, or mortality was not the outcome; these studies were only included in the meta-analysis of adverse effect. The hypothesis that publication bias might have affected the validity of the estimates was visually tested by a funnel plot–based approach.

Results

Characteristics of the studies

The workflow of the process of study selection is reported in Figure 1. A total of 42 articles were found in the search. Thirty-six of them were enrolled for analyzing the association with mortality of HCQ use in patients with COVID-19 (11 RCTs and 25 cohort studies), 11 were valuable for analyzing the association of HCQ+AZM and 13 for evaluating adverse effects of HCQ (Figure 1).

The main characteristics of the studies included in the meta-analyses are shown in Table 1. Data from 3 independent cohorts were extracted from the study of Kim et al. [46]; 7 articles were not published in peer reviewed journals; 4 observational studies reported unadjusted relative risks for the association between HCQ and mortality; 13 studies have been conducted in Europe, 17 in North America (Canada, USA or Mexico), 3 in Asia (China, Saudi Arabia) and three in other countries. The outcome considered was total mortality, with the exception of Geleris et al. [18] in which the authors used a combined endpoint formed by intubation or death; the mortality was intra-hospital, with the exception of the studies [28,33,35,36], in which death was all-cause, all-location. All studies included hospitalized patients, with the exclusion of three ones which included ambulatory [28,36] or non-hospitalized individuals [35]. In all studies the control group was formed by patients without HCQ exposure (HCQ or HCQ+AZM). All studies included adult men and women COVID-19 patients, with the exception of two RCT [9,35], that included a portion of individuals with uncertain positivity to Sars-CoV-2. A total of N = ‬50,048 ‬COVID-19 patients (including N = 8,709 from the 11 RCTs) were counted in the meta-analysis of HCQ, and N = 2,014 in the meta-analysis of HCQ+AZM. All but two cohort studies [24,48] (Supplementary Table 1) and three RCTs [9,38,49] (Supplementary Table 2) reported more than 80% of positive response to the Joanna Briggs Institute Critical Appraisal Checklists.

Table 1.

Characteristics of the studies included in the meta-analysis

Study Country Type of study N. of patients HCQ treatment (%) Mortality (%) Duration
(day, median)
Daily dose
(mg/day)
Total dose
(mg)
Abd-Elsalam S38 Egypt RCT 194 50.0 5.7 16 400 6800
Cavalcanti AB17 Brazil RCT 332 47.9 2.6 7 800 5600
Chen J40* China RCT 30 50.0 NA 5 400 2000
Chen L41* China RCT 30 60.0 0 10 400 4000
Chen Z42* China RCT 62 50.0 NA 5 400 2000
Dubée V43 France RCT 247 50.0 6.9 9 400 4000
Gonzalez JLB44 Mexico RCT 70 47.1 11.4 5 400 2400
Hernandez-Cardenas C45 Mexico RCT 214 52.6 39.3 10 400 4000
Horby P14 United Kingdom RCT 4716 33.1 25.6 10 800 9200
Lyngbakken MN49 Norway RCT 53 50.9 3.8 7 800 5600
Mitjà O29* Spain RCT 293 46.4 0 7 400 3600
Pan H9 30 Countries
worldwide
RCT 1853 51.1 11.1 11 800 10,000
Self WH33 USA RCT 479 50.5 10.4 5 400 2400
Skipper CP35 USA and Canada RCT 423 50 0.5 5 600 3800
Tang W50* China RCT 150 46.7 0 17 800 16,400
Ulrich RJ51 USA RCT 128 52.3 10.2 5 400 2400
Albani F15 Italy Cohort 816 25.9 25.7 6 400 2400
Arshad S11 USA Cohort 1611 74.6 18.1 5 400 2400
Ayerbe L16 Spain Cohort 2019 92.0 14.5 5 400 2800
Awad N39 USA Cohort 336 55.9 27.7 5 400 2400
Catteau L12 Belgium Cohort 8075 56.2 21.8 5 400 2400
Di Castelnuovo A7 Italy Cohort 3451 76.3 16.7 10 400 4400
Geleris J18 USA Cohort 1376 58.9 16.8 5 400 2800
Ip A19 USA Cohort 2256 84.8 22 5 400 2400
Kalligeros M20 USA Cohort 108 33.3 0.9 5 NA NA
Kim EJ46 USA Cohort A: 576 33.3 14.9 NA NA NA
B: 2816 50.0 21.7
C: 528 33.3 14.7
Lagier JC21 France Cohort 400 25.3 0.9 10 600 6000
Lamback BE47 Brazil Cohort 193 52.3§ 11.4 5 400 2400
Lammers AJJ22 The Netherlands Cohort 689 27.4 18 NA NA NA
Lauriola M23 Italy Cohort 80 21.3 38.7 10 600 6000
Lecronier M24 France Cohort 80 47.5 31.0 NA 400 NA
Lotfy SM48 Saudi Arabia Cohort 202 49.0 5.5 6 400 2800
Magagnoli J25 USA Cohort 277 51.1 17.3 5 400 2000
Mahévas M26 France Cohort 173 50.8 9.4 2 600 1200
Membrillo FJ27 Spain Cohort 166 73.5 28.9 5 400 2800
Mikami T28 USA Cohort 2820 73.7 21.7 5 NA NA
Paccoud O30 France Cohort 84 45.2 6.2 10 600 6000
Rosenberg ES31 USA Cohort 492 55.1 20.3 NA 800 NA
Sbidian E32 France Cohort 4415 14.1 21.4 10 400 4200
Singh S34 USA Cohort 1402 50 11.7 NA NA NA
Sulaiman T36 Saudi Arabia Cohort 5541 32.8 1.1 5 400 2400
Yu B37 China Cohort 550 8.7 44.9 8 400 3200

*Since in this study zero deaths in HCQ and/or control group has been observed, or mortality was not the outcome, it was included in the meta-analyses of adverse effect only. § prevalence of HCQ+AZM treatment. AZM: azithromycin; HCQ: hydroxychloroquine; RTC means randomized clinical trial; NA means not available

HCQ and death: RCT studies

Forest plot on the association between HCQ and mortality is reported in Figure 2, separately for RCTs and cohort studies.

Figure 2.

Figure 2.

Forest plot for association of hydroxychloroquine use with COVID-19 mortality (random effects)

HCQ means hydroxychloroquine; RCT means randomized clinical trial; SE means standard error. Data from 3 independent cohorts were extracted from the study of Kim et al. [46]

HCQ use was not associated with lower mortality after pooling data from 11 RCTs (pooled risk ratio: 1.08, 95%CI: 0.97 to 1.20; I2 = 0%). After removal of three RCTs [9,38,49] with an overall appraisal lower than 80%, the findings remain unchanged: pooled risk ratio 1.06, 95%CI: 0.95 to 1.19; I2 = 0%. The overall strength of evidence grade for the null association of HCQ use with total mortality observed in RCTs was judged high (Supplementary Table 3).

HCQ and death: cohort studies

Data pooling from 25 observational studies (27 cohorts) shows that the use of HCQ is associated with 20% lower mortality risk (pooled risk ratio: 0.80, 95%CI: 0.69 to 0.93; high level of heterogeneity: I2 = 80%, random effects; Figure 2). After the exclusion of 2 cohort studies [24,48] with an overall appraisal lower than 80% (Supplementary Table 1) the HCQ association with lower mortality remains unchanged (pooled risk ratio for cohort studies: 0.80, 95%CI: 0.68 to 0.94; I2 = 82%,) as it does after exclusion of the study by Geleris et al. [18] (which used a combined outcome of intubation and death; pooled risk ratio: 0.83, 95%CI: 0.72 to 0.95; I2 = 77%), or of the study by Membrillo et al. which appears as outlier (0.81, 95%CI: 0.70 to 0.94; I2 = 80%).

Since large heterogeneity in pooling data from cohort studies was found (I2 = 80%), as an alternative, we provide here a narrative description of results from individual observational studies, the same included in the quantitative review and whose characteristics are illustrated in Table 1. Namely, 8 studies reported a statistically significant association of HCQ use with lower mortality, with a relative risk ratio ranging from 0.07 to 0.70 among studies. Eight studies found that HCQ use was associated with a non-statistically significant reduced relative risk of mortality (range 0.62 to 0.99) and 11 cohorts reported a positive, non-statistically significant association with death (range 1.04 to 1.67). No studies found a positive, statistically significant association of HCQ use with mortality.

As the body of evidence has several deficiencies (Supplementary Table 3), the overall strength of evidence grade for the association of HCQ use with total mortality observed in cohort studies was judged low.

HCQ and death: subgroup analyses

Subgroup analyses according to the main features of primary studies are presented in Table 2, separately for RCT and cohort studies. In cohort studies, the association of HCQ with lower mortality was observed with very low differences in all subgroups, with the exception of dose grouping. The reduced mortality observed in cohort studies was in fact confined to studies that used a daily dose ≤400 mg (as estimated in days of treatment after the first, in which a higher (double for most) dose of drug was administered); pooling n = 5 studies which used more than 400 mg of HCQ daily resulted in an overall measure of association equal to 1.05 (95%CI: 0.73 to 1.53; Table 2 and Supplementary Figure 1A). Also, pooling studies which used more than 4,000 mg of HCQ during the entire phase of treatment set an overall measure of association equal to 0.86 (95%CI: 0.66 to 1.14) in comparison with studies which used ≤4,000 mg (0.67; 95%CI: 0.52 to 0.87) (Table 2 and Supplementary Figure 2A). Subgroup analysis according to both dosing (≤400 mg/d or >400 mg/d) and duration of treatment (≤5 days or >5 days) confirms the modification of effect by dosing of HCQ in cohort studies, with a maximum protection observed in studies with ≤400 mg/d and ≤5 days (0.65, 95%CI: 0.48 to 0.89; Supplementary Figure 3A) as opposed to studies with >400 mg/d and >5 days (0.98, 95%CI: 0.54 to 1.77; Supplementary Figure 3A).

Table 2.

Pooled analysis in subgroups of studies

  Cohort studies   RCT  
  N I2 Pooled RR*
[95% CI]
P
values§
N I2 Pooled RR*
[95% CI]
P
values§
ALL STUDIES 27# 80% 0.80 [0.69, 0.93]   11 0% 1.08 [0.97, 1.20]  
Peer-reviewed Studies                
Pre-printed studies 4 80% 0.68 [0.41, 1.12] 0.51 3 0% 0.73 [0.46, 1.15] 0.08
Peer-reviewed studies 23 81% 0.81 [0.69, 0.95]   8 0% 1.10 [0.99, 1.23]  
Adjustment                
Not adjusted 4 4% 1.02 [0.74, 1.41] 0.15 - - -  
Adjusted 23 82% 0.78 [0.67, 0.92]   11 0% 1.08 [0.97, 1.20]  
Sample size                
<1,000 COVID-19 patients 16 42% 0.93 [0.75, 1.15] 0.08 9 0% 0.89 [0.64, 1.23] 0.21
≥1,000 COVID-19 patients 11 90% 0.72 [0.58, 0.88]   2 0% 1.10 [0.99, 1.24]  
Mortality incidence in the sample                
≤20% 15 79% 0.80 [0.62, 1.03] 0.93 9 0% 1.08 [0.85, 1.36] 0.99
>20% 12 83% 0.81 [0.66, 0.99]   2 0% 1.08 [0.96, 1.22]  
HCQ treatment in the sample                
≤33.3% 10 54% 0.89 [0.69, 1.14] 0.02 1 - 1.09 [0.96, 1.23] 0.73
33.3–66.6% 11 74% 0.95 [0.79, 1.16]   10 0% 1.04 [0.84, 1.29]  
>66.6% 6 89% 0.52 [0.35, 0.75]   - - -  
HCQ treatment, duration                
≤ 5 days 12 85% 0.67 [0.52, 0.87] 0.35 4 0% 0.94 [0.56, 1.59] 0.61
> 5 days 8 46% 0.79 [0.63, 1.00]   7 0% 1.08 [0.97, 1.21]  
HCQ treatment, total dose                
≤4,000 mg 13 84% 0.67 [0.52, 0.87] 0.19 6 0% 0.85 [0.60, 1.21] 0.16
>4,000 mg 5 80% 0.86 [0.66, 1.14]   5 0% 1.10 [0.99, 1.24]  
HCQ treatment, daily dose                
≤400 mg 15 83% 0.69 [0.57, 0.85] 0.050 6 0% 0.85 [0.60, 1.19] 0.14
>400 mg 5 0% 1.05 [0.73, 1.53]   5 0% 1.11 [0.99, 1.24]  
Country       0.14       0.81
Asia 3 46% 0.48 [0.24, 0.95]   - - -  
Europe 12 59% 0.74 [0.62, 0.89]   3 2% 1.06 [0.89, 1.27]  
North America 12 86% 0.91 [0.72, 1.15]   5 0% 0.94 [0.56, 1.59]  
Others - - -   3 0% 1.18 [0.90, 1.56]  

*Relative risk for mortality in HCQ versus non HCQ patients. #Three independent cohorts have been included in the meta-analysis from the article of Kim et al.46 §for difference among subgroups.

HCQ means hydroxychloroquine; NA means Not Applicable; RCT means randomized clinical trial.

Although not statistically significant, a modification of effect by dosing was also observed in RCTs: the association with mortality for HCQ versus non HCQ use was 0.85 (95 CI%: 0.60 to 1.19) in 6 RCTs which used ≤400 mg/day and 1.11 (95 CI%: 0.99 to 1.24) in 5 RCTs which used >400 mg/day (Table 2 and Supplementary Figure 1B). Similar findings have been observed according to total dose of HCQ (Table 2 and Supplementary Figure 2B).

HCQ+AZM and death

Figure 3 reported random forest for 11 studies (one RCT) comparing HCQ+AZM. Use of the combination HCQ+AZM was associated with 25% lower mortality risk, with very large uncertainty (pooled risk ratio: 0.75, 95%CI: 0.51 to 1.10; P for testing of overall effect = 0.15; high level of heterogeneity: I2 = 90%, random effects; Figure 3).

Figure 3.

Figure 3.

Forest plot for association of hydroxychloroquine + azithromycin use with COVID-19 mortality (random effects)

AZM means azithromycin; HCQ means hydroxychloroquine.

By visual inspection of funnel plots (Supplementary Figure 4), we failed to observe any selection bias for both meta-analyses.

Adverse events of HCQ

Description of adverse effects as investigated in RCTs is reported in Supplementary table 4. Pooled analyses of the relationship between HCQ and incidence of adverse effects are reported in Supplementary Figure 5. HCQ use was associated with an increased risk of adverse effects of any type (pooled risk ratio: 1.95, 95%CI: 1.25 to 3.04; P for testing of overall effect = 0.003; Supplementary Figure 5A). On the contrary, patients treated with HCQ in RCTs showed a similar rate of serious adverse events, as that non-treated with HCQ (pooled risk ratio: 1.12, 95%CI: 0.88 to 1.44; P for testing of overall effect = 0.36; Supplementary Figure 5B).

Discussion

In a meta-analysis of 11 RCTs, the use of HCQ was not associated with mortality, but it was

associated with a 20% lower risk of mortality in a meta-analysis of 25 observational studies.

The potential for selection bias in observational studies is an important issue. The decision from the clinicians to utilize or not a drug may depend on comorbidities and baseline risk of the patient. However, in the pandemic, and in the absence of guidelines and specific anti COVID-19 drugs, allocation of HCQ in observational studies was not associated systematically with a lower or higher baseline risk profile. For example, in the CORIST study [7] patients receiving HCQ were more likely younger and less likely had ischemic heart disease, cancer or chronic kidney disease, but, on the contrary, they were more likely men and had higher levels of C-reactive protein. As a consequence, it is not clear if in that particular study HCQ patients were potentially at higher or lower risk of a negative prognosis. In attempting to account for baseline differences between patients who received HCQ and those who did not, we used the results for adjusted measure of association for each study, and this was possible for 23 out of 27 studies included in the meta-analysis of observational studies. After the exclusion of 4 unadjusted studies [21,24,39,48], the strength of the overall association of HCQ with mortality was unchanged. Although we attempted to control for potential confounding factors inherent to patient and clinical characteristics, it is possible that unmeasurable confounding still remains, and this may explain the different findings between the results of observational and RCT studies. However, it is hard to determine which are, if any, the unmeasured characteristics that have confused so strongly the association between HCQ and mortality in COVID-19 that was observed in observational studies. In fact, these features must be a) unmeasured in observational studies; b) associated with mortality in COVID-19 and c) associated with HCQ use, in a way that when the risky conditions are present, the clinicians tend systematically to avoid using HCQ. For example, HCQ is contraindicated in patients with cardiomyopathy but this condition has been mostly measured in observational studies and was not identified as a risk factor for mortality in COVID-19 patients.

The dissimilar findings between observational and RCTs we found might also be explained by differences in HCQ dosage [54]. Interestingly, we observed that the reduced mortality associated with HCQ treatment in observational studies was actually confined to studies that used a daily dose ≤400 mg, or a total dose ≤4,000 mg. Obviously these two conditions largely overlapped in studies, that we can now designate as ‘at low HCQ dosing’. Remarkably, 6 over 11 RCTs are in this category, but the others 5 are not, including the RECOVERY [14] and the SOLIDARITY study [9] which used 800 mg/day for 9 or 10 days (after the first), respectively, and a total dose of 9200 or 10,000 mg of HCQ (including the dose at first day) respectively, a very high dose regimen as compared to the rest of studies, particularly of the observational ones. Interestingly, though not statistically significant, the use of HCQ was associated with a 15% lower mortality in the six RCTs which used HCQ low doses, but a 10% higher mortality in the five RCTs which used high doses of the drug.

The possibility that HCQ reduced the risk of negative prognosis in COVID-19 patients when only administered at ‘low dose’ cannot be here undoubtedly proven, but it is a plausible hypothesis that may explain, at least in part, the different results between observational and RCT studies. More importantly, it might be useful in disentangling the debate on HCQ use in COVID-19. If our hypothesis of ‘low doses-short duration’ is correct, it follows that immunomodulation and inflammation occurs quite early after infection with SARS-CoV-2, as also suggested by the benefit of HCQ treatment in patients with higher C-reactive protein [7]. In this line, it is of interest that the studies with low doses and long duration of HCQ treatment also provided an overall negative association with mortality. This finding would also support the immunomodulatory hypothesis as potential mechanism of HCQ action [55] as it implies a cytokine rebound when the treatment is stopped. If this happens at a critical moment, it could worsen the patient clinical condition, thus vanishing the (potential) beneficial effect of HCQ [56].

High levels of HCQ administration were used in RCTs to maximize the antiviral activity of the drug that was considered to be the main mechanism of action of HCQ in this context. In some studies, the inverse association of HCQ with inpatient mortality was more evident in elderly, in patients who experienced a higher degree of COVID-19 severity or having elevated C-reactive protein levels [7], suggesting that the anti-inflammatory potential of HCQ may have had a more important role than its antiviral properties. HCQ, indeed, besides antiviral activity, may have both anti-inflammatory and anti-thrombotic effects [3]. This can justify its effect in reducing mortality risk, since Sars-Cov-2 can induce pulmonary microthrombi and coagulopathy, that are a possible cause of its severity [57,58] and the lack in preventing SARS-CoV-2 infection after exposure [59]. On the other end, national guidelines suggested to use HCQ 200 mg twice daily for 5–7 days probably to maintain a better risk/benefit profile hypothesizing that low doses could be more effective and safer. Indeed, non-sigmoidal, bell-shaped dose-response curves are possible with drugs having complex biological effects, multiple-binding sites or cellular and organ targets. On the other hand, anti SARS-2-CoV-2 activity of HCQ has been confirmed in Vero cells [60]. HCQ is also reported to reduce secretion of IFN-γ and IL-17 in activated Th1 and Th17 cells, respectively [61–62].

The concomitant use of azithromycin seems neither to increase nor decrease the effect, if any, of the HCQ since the combination of the two drugs was associated with a lower mortality risk to a very similar extent to that observed for HCQ alone; but the assumption is inconclusive because of the very large uncertainty in the findings.

A main concern with HCQ treatment have been its side effects, in particular, a severe cardiovascular toxicity. Indeed, HCQ can cause prolongation of the QT interval on electrocardiogram [61], which could be exacerbated by coadministration with azithromycin, widely prescribed as co-treatment in Covid-19 treatment. Our meta-analysis of data from RCTs, that allowed a proper evaluation of side effects, shows that use of HCQ was associated with an increase in side effects of any type, but not of major type, including cardiovascular events. This despite the high prevalence of cardiovascular disease in patients with COVID-19 or the high dose used in RCTs.

This meta-analysis has the strength of including all available studies that had not been included in previous meta-analyses, especially cohort studies [11,63]; findings on RCTs are in line with that from previous meta-analyses [63,64]. As a novelty, our meta-analysis considers modification of effect by dosing of HCQ. We recognize that the results obtained in observational or RCT studies were different and have discussed the possible implications of the difference observed. The pooled findings from cohort studies suffer of a high degree of heterogeneity, possibly depending from various factors, including setting, size of the study, dosing of HCQ and type of patients (in some studies HCQ was started right after the diagnosis but in others a large number of patients treated were already in intensive care).

Overall, the data of our meta-analysis suggest, though not proving, that a proportion of hospitalized COVID-19 patients might benefit of a treatment with low-dosage HCQ that, interestingly, is the same dosage as that currently used in some immuno-inflammatory and autoimmune diseases [65]. Research aimed to define the category of potentially HCQ-benefited patients may be worth being done in view of the persistent poverty of active anti-COVID-19 pharmacological treatments.

Conclusions

In conclusion, HCQ was not associated with decreased mortality in COVID-19 patients when RCTs studies were pooled (high level of certainty of evidence), but it was associated with 20% mortality reduction when cohort studies were combined (low level of certainty of evidence). The negative association with mortality was mainly apparent by pooling observational studies using lower doses of HCQ. Use of HCQ was not associated with severe adverse events.

Finding from cohort studies should be considered with caution because the overall strength of evidence grade was judged to be low.

At present, this is the largest comprehensive quantitative overview on the association of HCQ with mortality in COVID-19 patients, and our findings underscoring HCQ dosage effects might help disentangling the debate on HCQ use and encourage the planning of RCTs using low doses of HCQ (not necessarily with a short duration of the treatment) in hospitalized COVID-19 patients.

Supplementary Material

Supplemental Material

Acknowledgments

SC was the recipient of a Fondazione Umberto Veronesi Travel Grant.

Funding Statement

The authors have no funding to report.

Disclosure of potential conflicts of interest

No potential conflict of interest was reported by the author(s).

Source of support

none.

Data availability

All data generated or analysed during this study are included in this published article [and its supplementary information files].

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Availability of data and materials

All data generated or analysed during this study are included in this published article [and its supplementary information files] or in the primary published studies included in the meta-analysis.

Supplementary material

Supplemental data for this article can be accessed here

References

  • [1].Shukla AM, Wagle Shukla A.. Expanding horizons for clinical applications of chloroquine, hydroxychloroquine, and related structural analogues. Drugs Context. 2019. 2019 Nov 25;8:2019-9-1. Published doi: 10.7573/dic.2019-9-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Savarino A, Boelaert JR, Cassone A, et al. Effects of chloroquine on viral infections: an old drug against today’s diseases? Lancet Infect Dis. 2003;3(11):722–727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Quiros Roldan E, Biasiotto G, Magro P, et al. The possible mechanisms of action of 4-aminoquinolines (chloroquine/hydroxychloroquine) against Sars-Cov-2 infection (COVID-19): a role for iron homeostasis? Pharmacol Res. 2020;158:104904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Dowall SD, Bosworth A, Watson R, et al. Chloroquine inhibited Ebola virus replication in vitro but failed to protect against infection and disease in the in vivo guinea pig model. J Gen Virol. 2015;96(12):3484–3492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Savarino A, Shytaj IL. Chloroquine and beyond: exploring anti-rheumatic drugs to reduce immune hyperactivation in HIV/AIDS. Retrovirology. 2015. 2015 Jun 18;12(1):51. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].De Wilde AH, Jochmans D, Posthuma CC, et al. Screening of an FDA-approved compound library identifies four small-molecule inhibitors of Middle East respiratory syndrome coronavirus replication in cell culture. Antimicrob Agents Chemother. 2014;58(8):4875–4884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Castelnuovo AD, Costanzo S, Antinori A, COVID-19 RISK and Treatments (CORIST) Collaboration . Use of hydroxychloroquine in hospitalised COVID-19 patients is associated with reduced mortality: findings from the observational multicentre Italian CORIST study. Eur J Intern Med. 2020;82:38–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Mehra MR, Desai SS, Ruschitzka F, et al. Hydroxychloroquine or chloroquine with or without a macrolide for treatment of COVID-19: a multinational registry analysis. Lancet. 2020. May 22. Retracted article. doi: 10.1016/S0140-6736(20)31180-6 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [9].Pan H, Peto R, Henao-Restrepo AM, et al. WHO Solidarity Trial Consortium, . Repurposed antiviral drugs for Covid-19 - interim WHO solidarity trial results. N Engl J Med. 2021;384(6):497–511. . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Mehra MR, Ruschitzka F, Patel AN. Retraction-Hydroxychloroquine or chloroquine with or without a macrolide for treatment of COVID-19: a multinational registry analysis [retraction of: lancet. 2020 May 22]. Lancet. 2020;395(10240):1820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Fiolet T, Guihur A, Rebeaud ME, et al. Effect of hydroxychloroquine with or without azithromycin on the mortality of coronavirus disease 2019 (COVID-19) patients: a systematic review and meta-analysis. Clin Microbiol Infect. 2021;27(1):19–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Arshad S, Kilgore P, Chaudhry ZS, et al. Treatment with hydroxychloroquine, azithromycin, and combination in patients hospitalized with COVID-19. Int J Infect Dis. 2020;97:396–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Catteau L, Dauby N, Montourcy M, et al. Low-dose hydroxychloroquine therapy and mortality in hospitalised patients with COVID-19: a nationwide observational study of 8075 participants. Int J Antimicrob Agents. 2020;56(4):106144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Horby P, Mafham M, Linsell L, et al. RECOVERY Collaborative Group . Effect of hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med. 2020;383(21):2030–2040. . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Albani F, Fusina F, Giovannini A, et al. Impact of Azithromycin and/or Hydroxychloroquine on Hospital Mortality in COVID-19. J Clin Med. 2020. 2020 Aug 30;9(9):2800. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Ayerbe L, Risco-Risco C, Ayis S. The association of treatment with hydroxychloroquine and hospital mortality in COVID-19 patients. Intern Emerg Med. 2020;15(8):1501–1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Cavalcanti AB, Zampieri FG, Rosa RG, et al. Hydroxychloroquine with or without Azithromycin in Mild-to-Moderate Covid-19 [published correction appears in N Engl J Med. 2020 Nov 19;383(21):e119]. N Engl J Med. 2020;383(21):2041–2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Geleris J, Sun Y, Platt J, et al. Observational study of Hydroxychloroquine in hospitalized patients with Covid-19. N Engl J Med. 2020;382(25):2411–2418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Ip A, Berry DA, Hansen E, et al. Hydroxychloroquine and tocilizumab therapy in COVID-19 patients-An observational study. PLoS One. 2020. 2020 Aug 13;15(8):e0237693. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Kalligeros M, Shehadeh F, Atalla E, et al. Hydroxychloroquine use in hospitalised patients with COVID-19: an observational matched cohort study. J Glob Antimicrob Resist. 2020;22:842–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Lagier JC, Million M, Gautret P, et al. Outcomes of 3,737 COVID-19 patients treated with hydroxychloroquine/azithromycin and other regimens in Marseille, France: a retrospective analysis. Travel Med Infect Dis. 2020;36:101791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Lammers AJJ, Brohet RM, Theunissen REP, et al. Early hydroxychloroquine but not chloroquine use reduces ICU admission in COVID-19 patients. Int J Infect Dis. 2020;101:283–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Lauriola M, Pani A, Ippoliti G, et al. Effect of combination therapy of Hydroxychloroquine and Azithromycin on mortality in patients with COVID-19. Clin Transl Sci. 2020;13(6):1071–1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Lecronier M, Beurton A, Burrel S, et al. Comparison of hydroxychloroquine, lopinavir/ ritonavir,and standard of care in critically ill patients with SARS-CoV-2 pneumonia: an opportunistic retrospective analysis. Crit Care. 2020. 2020 Jul 11;24(1):418. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Magagnoli J, Narendran S, Pereira F, et al. Outcomes of Hydroxychloroquine usage in United States veterans hospitalized with COVID-19. Med (N Y). 2020;1(1):114–127.e3. . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Mahévas M, Tran VT, Roumier M, et al. Clinical efficacy of hydroxychloroquine in patients with covid-19 pneumonia who require oxygen: observational comparative study using routine care data [published correction appears in BMJ. 2020 Jun 18;369:m2328]. BMJ. Published. 2020;369:m1844. 2020 May 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Membrillo De Novales FJ, Ramírez-Olivencia G, Estébanez M, et al. Early Hydroxychloroquine is associated with an increase of survival in COVID-19 patients: an observational study. Preprints. 2020;2020050057. DOI: 10.20944/preprints202005.0057.v1. [DOI] [Google Scholar]
  • [28].Mikami T, Miyashita H, Yamada T, et al. Risk factors for mortality in patients with COVID-19 in New York City. J Gen Intern Med. 2021;36(1):17–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Mitjà O, Corbacho-Monné M, Ubals M, et al. Hydroxychloroquine for early treatment of adults with mild Covid-19: a Randomized-Controlled trial [published online ahead of print, 2020 Jul 16]. Clin Infect Dis. 2020;ciaa1009. DOI: 10.1093/cid/ciaa1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Paccoud O, Tubach F, Baptiste A, et al. Compassionate use of hydroxychloroquine in clinical practice for patients with mild to severe Covid-19 in a French university hospital [published online ahead of print, 2020 Jun 18]. Clin Infect Dis. 2020;ciaa791. doi: 10.1093/cid/ciaa791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Rosenberg ES, Dufort EM, Udo T, et al. Association of treatment with Hydroxychloroquine or Azithromycin with in-Hospital mortality in patients with COVID-19 in New York State. JAMA. 2020;323(24):2493–2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Sbidian E, Josse J, Lemaitre G, et al. Hydroxychloroquine with or without azithromycin and in-hospital mortality or discharge in patients hospitalized for COVID-19 infection: a cohort study of 4,642 in-patients in France. medRxiv. 2020. June 16;20132597. doi: 10.1101/2020.06.16.20132597. [DOI] [Google Scholar]
  • [33].Self WH, Semler MW, Leither LM, et al. Effect of Hydroxychloroquine on clinical status at 14 days in hospitalized patients with COVID-19: a randomized clinical trial. JAMA. 2020;324(21):2165–2176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Singh S, Khan A, Chowdhry M, et al. Outcomes of Hydroxychloroquine treatment among hospitalized COVID-19 patients in the United States- Real-World evidence from a federated electronic medical record network. medRxiv. 2020. May 12; 20099028. doi: 10.1101/2020.05.12.20099028. [DOI] [Google Scholar]
  • [35].Skipper CP, Pastick KA, Engen NW, et al. Hydroxychloroquine in nonhospitalized adults with early COVID-19: a randomized trial. Ann Intern Med. 2020;173(8):623–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Sulaiman T, Mohana A, Alawdah L, et al. The effect of early Hydroxychloroquine-based Therapy in COVID-19 patients in ambulatory care settings: a nationwide prospective cohort study. medRxiv. 2020. September 09;20184143. doi: 10.1101/2020.09.09.20184143. [DOI] [Google Scholar]
  • [37].Yu B, Li C, Chen P, et al. Low dose of hydroxychloroquine reduces fatality of critically ill patients with COVID-19 [published correction appears in sci China life sci. Sci China Life Sci. 2020;63(10):1515–1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Abd-Elsalam S, Esmail ES, Khalaf M, et al. Hydroxychloroquine in the Treatment of COVID-19: a multicenter randomized controlled study. Am J Trop Med Hyg. 2020;103(4):1635–1639. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [39].Awad N, Schiller DS, Fulman M, et al. Impact of hydroxychloroquine on disease progression and ICU admissions in patients with SARS-CoV-2 infection [published online ahead of print, 2021 Feb 18]. Am J Health Syst Pharm. 2021;78(8):zxab056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Chen J, Liu D, Liu L, et al. A pilot study of hydroxychloroquine in treatment of patients with common coronavirus disease-19 (COVID-19). Zhejiang Da Xue Xue Bao Yi Xue Ban. 2020;49(2):215–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Chen L, Zhang ZY, Fu JG, et al. Efficacy and safety of chloroquine or hydroxychloroquine in moderate type of COVID-19: a prospective open-label randomized controlled study. medRxiv. 2020. June 19;20136093. doi: 10.1101/2020.06.19.20136093. [DOI] [Google Scholar]
  • [42].Chen Z, Hu J, Zhang Z, et al. Efficacy of hydroxychloroquine in patients with COVID-19: results of a randomized clinical trial. medRxiv. 2020. March 22;20040758. doi: 10.1101/2020.03.22.20040758. [DOI] [Google Scholar]
  • [43].Dubée V, Roy PM, Vielle B, et al. A placebo-controlled double blind trial of hydroxychloroquine in mild-to-moderate COVID-19. medRxiv. 2020. October 19;20214940. doi: 10.1101/2020.10.19.20214940. [DOI] [Google Scholar]
  • [44].Gonzalez JLB, González Gámez M, Mendoza Enciso EA, et al. Efficacy and safety of Ivermectin and Hydroxychloroquine in patients with severe COVID-19. A randomized controlled trial. medRxiv. 2021;2:18.21252037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Hernandez-Cardenas C, Thirion-Romero I, Rivera-Martinez NE, et al. Hydroxychloroquine for the treatment of severe respiratory infection by covid-19: a randomized controlled trial. medRxiv. 2021. 02.01.21250371. doi: 10.1101/2021.02.01.21250371 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Kim EJ, Coppa K, Hirsch JS, et al. Examination of patient characteristics and hydroxychloroquine use based on the US food and drug administration’s recommendation: a cross-sectional analysis in New York. BMJ Open. 2021. 2021 Feb 8;11(2):e042965. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Lamback EB, Oliveira MA, Haddad AF, et al. Hydroxychloroquine with azithromycin in patients hospitalized for mild and moderate COVID-19 [published online ahead of print, 2021 Feb 19]. Braz J Infect Dis. 2021;25(2):101549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Lotfy SM, Abbas A, Shouman W. Use of Hydroxychloroquine in patients with COVID-19: a retrospective observational study. Turk Thorac J. 2021;22(1):62–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Lyngbakken MN, Berdal JE, Eskesen A, et al. A pragmatic randomized controlled trial reports lack of efficacy of hydroxychloroquine on coronavirus disease 2019 viral kinetics. Nat Commun. 2020. 2020 Oct 20;11(1):5284. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Tang W, Cao Z, Han M, et al. Hydroxychloroquine in patients with mainly mild to moderate coronavirus disease 2019: open label, randomised controlled trial. BMJ. Published. 2020;369:m1849. 2020 May 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Ulrich RJ, Troxel AB, Carmody E, et al. Treating COVID-19 with Hydroxychloroquine (TEACH): a Multicenter, Double-blind randomized controlled trial in hospitalized patients. Open Forum Infect Dis. 2020. 2020 Sep 23;7(10):ofaa446. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Moola S, Munn Z, Tufanaru C, et al. 7: systematic reviews of etiology and risk - Joanna briggs institute reviewers’ manual. Joanna Briggs Institute Reviewer’s Manual. 2017
  • [53].Berkman ND, Lohr KN, Ansari M, et al. Grading the strength of a body of evidence when assessing health care interventions for the effective health care program of the agency for healthcare research and quality: an update. Methods Guide for Comparative Effectiveness Reviews (Prepared by the RTI-UNC Evidence-based Practice Center under Contract No. 290-2007-10056-I). AHRQ Publication No. 13(14)-EHC130-EF. Rockville, MD: Agency for Healthcare Research and Quality. November 2013 . www.effectivehealthcare.ahrq.gov/reports/final.cfm [PubMed]
  • [54].Ippolito MM, Flexner C. Dose optimization of Hydroxychloroquine for coronavirus infection 2019: do Blood concentrations matter? Clin Infect Dis. 2020;71(11):2965–2967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Plantone D, Koudriavtseva T. Current and future use of Chloroquine and Hydroxychloroquine in infectious, immune, neoplastic, and neurological diseases: a Mini-Review. Clin Drug Investig. 2018;38(8):653–671. [DOI] [PubMed] [Google Scholar]
  • [56].Wang C, Xie J, Zhao L, et al. Aveolar macrophage activation and cytokine storm in the pathogenesis of severe COVID-19. Research Square. 26 Mar, 2020. doi: 10.21203/rs.3.rs-19346/v1 [DOI]
  • [57].McGonagle D, Sharif K, O’Regan A, et al. The role of cytokines including interleukin-6 in COVID-19 induced pneumonia and macrophage activation syndrome-Like disease. Autoimmun Rev. 2020;19(6):102537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Thachil J. The versatile heparin in COVID-19. J Thromb Haemost. 2020;18(5):1020–1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Boulware DR, Pullen MF, Bangdiwala AS, et al. A randomized trial of Hydroxychloroquine as Postexposure Prophylaxis for Covid-19. N Engl J Med. 2020;383(6):517–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Liu J, Cao R, Xu M, et al. Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro. Cell Discov. 2020. 2020 Mar 18;6(1):16. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Schmidt RL, Jutz S, Goldhahn K, et al. Chloroquine inhibits human CD4+ T-cell activation by AP-1 signaling modulation. Sci Rep. 2017. 2017 Feb 7;7(1):42191. . Published. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Jankelson L, Karam G, Becker ML, et al. QT prolongation, torsades de pointes, and sudden death with short courses of chloroquine or hydroxychloroquine as used in COVID-19: a systematic review. Heart Rhythm. 2020;17(9):1472–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Kashour Z, Riaz M, Garbati MA, et al. Efficacy of chloroquine or hydroxychloroquine in COVID-19 patients: a systematic review and meta-analysis. J Antimicrob Chemother. 2021;76(1):30–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Kashour Z, Kashour T, Gerberi D, et al. . Mortality, viral clearance, and other clinical outcomes of hydroxychloroquine in COVID-19 Patients: a Systematic Review and Meta-Analysis of Randomized Controlled Trials. Clin Transl Sci. 2021Feb19. doi: 10.1111/cts.13001 Epub ahead of print. PMID: 33606894; PMCID: PMC8013604 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Rainsford KD, Parke AL, Clifford-Rashotte M, et al. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology. 2015. Oct;23(5):231–269. . Epub 2015 Aug 6. PMID: 26246395. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Material

Data Availability Statement

All data generated or analysed during this study are included in this published article [and its supplementary information files].

All data generated or analysed during this study are included in this published article [and its supplementary information files] or in the primary published studies included in the meta-analysis.


Articles from Pathogens and Global Health are provided here courtesy of Taylor & Francis

RESOURCES