Beneficial and harmful outcomes of tocilizumab in severe COVID-19: a systematic review and meta-analysis ======================================================================================================== * Manuel Rubio-Rivas * Jose María Mora-Luján * Abelardo Montero * Narcís A. Homs * Jordi Rello * Xavier Corbella ## ABSTRACT **Objectives** Pending for randomized control trials, the use of tocilizumab (TCZ) in COVID-19 remains controversial. We performed a systematic review and meta-analysis to investigate the effect on clinical outcomes of TCZ to treat severe COVID-19. **Methods** From 1 January to 21 August 2020, we searched PubMed (via MEDLINE), Scopus, and medRxiv repository databases for observational studies in any language reporting efficacy and safety of TCZ use in hospitalized adults with COVID-19. Independent and dually data extraction and quality assessment were performed. **Results** Of 57 eligible studies, 27 controlled and 30 not. The overall included patients were 8,128: 4,021 treated with TCZ, in addition to standard of care (SOC), and 4,107 only receiving SOC. The pooled mortality was lower in the TCZ-group, with a relative risk (RR) of 0.73 (95%CI 0.57-0.93; p=0.010). TCZ-treated patients were transferred to the intensive care unit (ICU) in a higher proportion, but ICU mortality was lower than in the control group. Conversely, a higher proportion of TCZ-treated patients developed secondary infections after TCZ use. **Conclusions** TCZ seems beneficial in preventing in-hospital mortality in severe, non-critically ill COVID-19 patients. However, patients receiving TCZ appear to be at higher risk for secondary infections, especially those admitted to ICU. Keywords * Coronavirus * SARS-CoV-2 * COVID-19 * Tocilizumab * Meta-analysis ## INTRODUCTION Since early 2020, when the SARS-CoV-2 pandemic hit the world, a variety of treatments have been suggested for COVID-19 [1-5]. However, to date, only remdesivir [4] and dexamethasone [5] have demonstrated evidence-based efficacy on randomized, controlled clinical trials (RCTs). This double strategy combining antiviral and immunomodulatory therapy is in accordance with the two pathological mechanisms that appear to coexist in the disease; the first triggered by the virus itself and the second by the cytokine storm and systemic dysregulated host-immune hyperinflammatory response [6]. While the pandemic continues to spread globally, a worrying 15% of patients continue to transit into the most severe stage of the disease, requiring hospitalization or intensive care unit (ICU) admission. This advanced clinical-stage presents as severe pulmonary injury and multi-organ failure, causing fatality in nearly half of cases, resembling complications from CAR T cell therapy [7]. Among other pro-inflammatory cytokines, IL-6 plays a part in innate immunity, but excessive production by the host facing SARS-CoV-2 is detrimental [8-10]. Accordingly, the use of immunomodulatory agents such as tocilizumab (TCZ), a monoclonal antibody to the recombinant human IL-6 receptor, was initially reported as successful among 21 patients in China [11]. Since then, an emerging number of observational studies from America and Europe have been published or registered assessing the effect of TCZ in severe COVID-19 [12-67]. In most of them, the authors report an association between earlier use of TCZ and reduced mortality; however, interpretation of these results is limited because several of them did not describe a comparison group. Conversely, given preliminary results from the industry-sponsored Phase 3 COVACTA trial (ClinicalTrials.gov Identifier [NCT04320615](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04320615&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom)), the COVID-19 Treatment Guidelines Panel by the National Institute of Health, has taken a position against the use of TCZ [68]. This RCT is the first global, randomized, double-blind, placebo-controlled phase III trial investigating TCZ in hospitalized patients with severe COVID-19, but it failed to demonstrate improvement in clinical status as the primary endpoint, or several key secondary outcomes such as 4-week mortality [69]. In the current emergency, while waiting for additional data from RCTs, the fact that most institutions and physicians worldwide are still tackling COVID-19 based only on real-world reported data, prompted us the present review. Our aim was to summarize the updated results from available observational studies on the effect of TCZ on clinical outcomes in hospitalized patients with COVID-19. ## METHODS This systematic review and meta-analysis followed the guidance of the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement [70]. The protocol was published in the National Institute for Health Research international register of systematic reviews (PROSPERO); registration number CRD42020204934. A clinical question under the PICO framework format (Population-Intervention-Comparison-Outcome) was created (Table 1). View this table: [Table 1.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/T1) Table 1. PICO framework format ### Data Sources and Searches The search strategy was developed by three investigators (M.R-R., J.R., X.C.), which was revised and approved by the other investigators (J.M.M-L., A.M, N.A.H.). We searched the following databases from 1 January to 21 August 2020: MEDLINE database through the PubMed search engine, Scopus, and the medRxiv repository, using the terms “COVID-19” [MesH]) AND “Tocilizumab” [MesH]. ### Study Eligibility Criteria Full-text observational studies in any language reporting beneficial or harmful outcomes from the use of TCZ in adults hospitalized with COVID-19 were included. Two investigators (M.R-R., J.M.M-L.) independently screened each record title and abstract for potential inclusion. Restriction of publication type was manually applied: secondary analyses of previously reported trials, protocols, abstracts-only and experimental studies were excluded. Potentially relevant articles were retrieved for full-text review. Two investigators (M.R-R., A.M.) read the full text of the abstracts selected. Discrepancies were resolved through discussion or by a third investigator (X.C.). Publications were included if they met all the following criteria: 1) the study reported data on adults ≥18 years-old with COVID-19, diagnosed by polymerase chain reaction (PCR), admitted hospital-wide or in ICUs; 2) the study design was an observational investigation providing real-world original data on TCZ use in COVID-19, either intravenous or subcutaneous; and 3) the study data collection finished after 1 January 2020. Those studies reported being “case-control studies”, in which subjects from the control group also presented COVID-19, just as those from the TCZ group, were also included in the present review. Studies focusing on a sole subgroup of patients (e.g. renal transplant recipients) were excluded. A careful revision was also performed of patients’ origins included in studies from the same country/hospital to avoid overlapping data, and only the latest and largest study was selected. The search was completed by the bibliography review of every paper selected for full-text examination. ### Data Extraction and Quality Assessment Two investigators (M.R-R., J.M.M-L.) independently abstracted the following details: study characteristics, including setting; intervention or exposure characteristics, including medication dose and duration; patient characteristics, including the severity of disease; and outcomes, including mortality, admission to ICU, adverse events such as secondary infections, and length of hospital stay. Discrepancies were resolved by discussion in consultation with a third investigator (X.C.). Quality assessment was performed by two investigators (M.R-R., N.A.H) using the Newcastle-Ottawa Scale (NOS) for observational studies [71]. In case of disagreement, a third author (J.R.) independently determined the quality assessments. ### Data Analysis Categorical variables were described as absolute numbers and percentages. When the number of events and the sample size was small (and followed a Poisson distribution), confidence intervals were estimated using Wilson’s method [72-74]. We carried out a meta-analysis of the pooled mortality ratio by including all comprised studies. Those studies with a control group were also meta-analyzed to assess the relative risk (RR) of mortality in TCZ-treated patients vs. those non-TCZ treated (RR of 1). The inverse variance-weighted method was initially performed using a fixed-effects model. Heterogeneity between studies was assessed using the Q statistic. The percentage of variability between studies by the Higgins I2 parameter and between-study variability was measured by the Tau2 parameter and, when confirmed (p<0.05), the analysis was completed by using the random-effects model [75]. Studies with 0 events in the only arm (uncontrolled studies) or both arms (controlled studies) were not included in the meta-analysis. Forest plots were depicted accordingly. Publication bias was assessed using the Egger method [76]. Statistical analysis was performed by IBM SPSS Statistics for Windows, Version 26.0. Armonk, NY: IBM Corp. ## RESULTS A total of 781 articles were identified in our search. Of these, 81 qualified for full-text review following title and abstract screening, of which 57 [11-67] were included in the analysis. The PRISMA flow diagram is detailed in Figure 1. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/09/26/2020.09.05.20188912/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/F1) Figure 1. PRISMA 2009 Flow Diagram ### Study characteristics The majority of included studies were carried out in different hospitals in high-income countries in America and Europe, such as the US [13, 18, 22, 28, 31, 34, 35, 48, 49, 53-60, 66], Italy [14, 15, 17, 19, 20, 41, 44-47, 50, 51, 63, 65], Spain [21, 26, 27, 32, 33, 39, 40, 42, 43], and France [25, 29, 30, 61, 62, 64]. A lesser number were conducted in China [11, 12]. The distribution of the studies worldwide is shown in Figure 2. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/09/26/2020.09.05.20188912/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/F2) Figure 2. Worldwide Distribution of the Included Studies. ### Search results Of the total of 57 studies included, 12 were prospective and 45, retrospective. In 30 of these cohort investigations, a control group was not described [10-38] and 27 added a comparison group [40-67]. The overall results provided data from 8,128 hospitalized patients with COVID-19: 4,021 TCZ-treated, in addition to standard of care (SOC) (including 711 patients admitted to ICU), and 4,107 only receiving SOC (including 694 patients admitted to ICU). SOC was basically antiviral therapy (remdesivir, lopinavir/ritonavir), antimalarials (hydroxychloroquine), or azythromicin. Characteristics of patients in the TCZ-group vs. the control group are detailed in Tables 2-7. Of the TCZ group, 2,645 (65.8%) were men with a mean age of 61.8 (SD 6.1) and median age 62.6 [range 59-65], according to the data provided. TCZ was given as a single dose in 2,030/2,952 patients (68.8%), and in 922 (31.2%), as two or more doses. View this table: [Table 2.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/T2) Table 2. Non-controlled studies. General data. View this table: [Table 3.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/T3) Table 3. Controlled studies. General data. View this table: [Table 4.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/T4) Table 4. Non-controlled studies. Drugs and mortality. View this table: [Table 5.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/T5) Table 5. Controlled studies. Drugs and mortality. View this table: [Table 6.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/T6) Table 6. Non-controlled studies. Length of hospital stay, Mortality ICU vs. ward and secondary infections. View this table: [Table 7.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/T7) Table 7. Controlled studies. Length of hospital stay, Mortality ICU vs. ward and secondary infections. Concomitantly to TCZ use, additional treatment with steroids was given in 1,560/3,073 patients in the TCZ-group (50.8%) vs. 592/2,733 (21.7%) in the control group (p<0.001). Comparing both groups, remdesivir was used in 37/3,511 (1.05%) vs. 23/3,945 (0.58%) (p=0.023) patients. Finally, the administration of TCZ has prescribed a median of 10 days [range 9-11] after the onset of COVID-19 symptoms, in those studies in which this data was provided. The median follow-up of the overall cohort was 10.3 days [range 12-19]. ### Mortality After applying the random-effects model, hospital-wide (including ICUs) pooled mortality of patients with COVID-19 treated with TCZ was 19.2% (95%CI 16.4-22.5) (I2=83.6% Q=305.3 tau2=0.23 p<0.001). Egger’s method A=-3.354 p<0.001 (Figure 3). In the control group, overall mortality was 27.4% (95%CI 21.1-35.6%) (I2=95.9% Q=569.1 tau2=0.38 p<0.001). These differences between the TCZ-group and the control group achieved statistical significance (p<0.001). The RR of mortality in the TCZ-group was 0.73 (95%CI 0.57-0.93; p=0.010) (I2=77.7% Q=107.4 tau2=0.24 p<0.001). Egger’s method A=- 0.712 p=0.380 (Figure 4). The number needed to treat (NNT) to avoid one death was 20. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/09/26/2020.09.05.20188912/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/F3) Figure 3. Mortality (percentage) in COVID-19 patients receiving TCZ. Forest plot. ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/09/26/2020.09.05.20188912/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2020/09/26/2020.09.05.20188912/F4) Figure 4. Mortality (RR) in the TCZ-group vs. the control group. Forest plot. When the analysis was restricted to high-quality controlled observational studies, excluding those non-controlled, those with <20 patients, or those showing NOS <7, the pooled mortality in the TCZ-group was 17.7% (95%CI 13.9-22.6) (I2=85% Q=168 tau2=0.295 p<0.001). Egger’s method A=-3.727 p<0.001. The RR of death in the TCZ-group vs. the control group was 0.58 (95%CI 0.40-0.85) (I2=79.7% Q=108.3 tau2=0.60 p=0.004). Egger’s method A=-1.511 p=0.153. #### Overall Mortality in hospital wards The pooled mortality of COVID-19 patients receiving TCZ in conventional wards was 17% (95%CI 13.9-20.8) (I2=58.5% Q=50.6 tau2=0.108 p<0.001). Egger’s method A=-2.082 p<0.001. In contrast, mortality in the control group after being admitted for COVID-19 in conventional wards was 23.8% (95%CI 14.8-38.4) (I2=90.3% Q=51.8 tau2=0.314 p<0.001). Egger’s method A=5.433 p=0.044. These differences did not achieve statistical significance (p=0.261). The RR of mortality in hospital wards in the TCZ-group vs. the control group was 0.64 (95%CI 0.26-1.54; p=0.314) (I2=80.8% Q=26 tau2=0.91 p=0.314). Egger’s method A=-3.576 p=0.005. When analysis was restricted to high-quality controlled studies, the pooled mortality in hospital wards in the TCZ-group was 13.3% (95%CI 8.7-20.4) (I2=76.2% Q=25 tau2=0.235 p<0.001). Egger’s method A=-2.653 p=0.074. #### Overall Mortality in ICUs The pooled mortality of COVID-19 patients receiving TCZ once admitted to ICU was 31.7% (95%CI 24.7-40.8) (I2=82.6% Q=97.6 tau2=0.185 p<0.001). Egger’s method A=-2.995 p<0.001. When compared, mortality of ICU control group patients was 39.1% (95%CI 29.1-52-4) (I2=91.6% Q=59.4 tau2=0.101 p<0.001). Egger’s method A=-1.766 p=0.290. These differences achieved statistical significance (p<0.001). The RR for mortality in those ICU patients receiving TCZ vs. SOC was 0.78 (95%CI 0.44-1.35; p=0.369) (I2=65.4% Q=14.4 tau2=0.29 p=0.639). Egger’s method A=0.693 p=0.727. The NNT in ICU to avoid one death was 9. When the analysis was restricted to high-quality controlled studies, the pooled mortality in the ICU in the TCZ-group was 29.3% (95%CI 19.4-44.2) (I2=89.1% Q=55 tau2=0.235 p<0.001). Egger’s method A=-4.564 p=0.009. #### Mortality in the TCZ-group receiving additional corticosteroids Concomitantly to TCZ use, additional treatment with steroids was given in 1,560/3,073 patients in the TCZ-group (50.8%) vs. 592/2,733 (21.7%) in the control group (p<0.001). Since outcomes were not reported separately in most of the included studies, differences in mortality could only be compared between those studies in which steroids were not prescribed in addition of TCZ use and those in which 100% of TCZ-treated patients received concomitant therapy with steroids. The pooled mortality of TCZ patients without corticosteroids was 10.8% (95%CI 6.3-18.5) (I2=91% Q=136.3 tau2=0.820 p<0.001). Egger’s method A=-4.438 p=0.006. ***Alternatively,*** the pooled mortality of TCZ patients with corticosteroids was 16.3% (95%CI 12.8-20.8) (I2=35.9% Q=6.2 tau2=0.046 p<0.001). Egger’s method A=-0.342 p=0.895. #### Mortality in the TCZ-group receiving early vs. late TCZ administration Hospital-wide (including ICUs) pooled mortality in TCZ-treated patients in whom TCZ was early administered (<10 days from symptoms onset) was 15.9% (95%CI 10.9-23) (I2=74.9% Q=31.9 tau2=0.222 p<0.001). Egger’s method A=-2.306 p=0.239. Hospital-wide (including ICUs) pooled mortality in patients with COVID-19 illness in whom TCZ was administered later (>10 days) was 23.3% (95%CI 17.9-30.3) (I2=75.5% Q=36.8 tau2=0.115 p<0.001). Egger’s method A=-2.983 p=0.021. Differences between groups did not achieve statistical significance (p=0.252). When the analysis was restricted to high-quality controlled studies, the pooled mortality after early TCZ was 11% (95%CI 8.4-14.3) (I2=42.4% Q=5.2 tau2=0.057 p<0.001). Egger’s method A=-2.824 p=0.372. Alternatively, the pooled mortality after late TCZ was 22.6% (95%CI 16.4-31.1) (I2=3.4% Q=2.1 tau2=0.004 p<0.001). Egger’s method A=-2.756 p=0.034. ### Risk of ICU admission In patients with COVID-19 initially admitted to hospital wards, the pooled ICU admission rate after TCZ administration was 17.1% (95%CI 11.5-25.5) (I2=90% Q=149.5 tau2=0.51 p<0.001). Egger’s method A=-3.272 p=0.015. Conversely, the risk for ICU admission in the control group, initially admitted outside the ICUs, was 9.5% (95%CI 2.9-31.2) (I2=96.2% Q=78.8 tau2=1.37 p<0.001). Egger’s method A=-1.315 p=0.886. These differences achieved statistical significance (p<0.001). Accordingly, in the subset of patients initially admitted outside the ICUs, those receiving TCZ showed a RR of ICU admission of 1.49 (95%CI 0.30-7.34; p=0.621) (I2=93% Q=43 tau2=2.35 p=0.62). Egger’s method A=-4.026 p=0.435. When the analysis was restricted to high-quality controlled studies, the pooled risk of ICU admission after receiving TCZ at the ward was 15.6% (95%CI 11.8-20.9) (I2=22.4% Q=3 tau2=0.022 p<0.001). Egger’s method A=-1.839 p=0.113. ### Safety The pooled rate of reported secondary viral, bacterial or opportunistic fungal infections in those patients with COVID-19 treated with TCZ was 18.9% (95%CI 14.5-24.8) (I2=88.1% Q=218.8 tau2=0.391 p<0.001). Egger’s method A=-3.852 p=0.001. The RR of secondary infections in TCZ-treated vs. the control group was 1.47 (95%CI 0.99-2.19; p=0.058) (I2=66.1% Q=38.3 tau2=0.34 p=0.058). Egger’s method A=0.039 p=0.977 When the analysis was restricted to high-quality controlled studies, the pooled percentage of secondary infections was 20.7% (95%CI 14.6-29.2) (I2=88.2% Q=118.5 tau2=0.351 p<0.001). Egger’s method A=-4.128 p=0.003. ### Length of hospital stay Among survivors, the length of hospital stay in the TCZ-group was a median of 15.3 days [range 12.4-19.4] vs. 14 days [range 9-20] in the control group. These differences were not statistically significant (p=0.953). ## DISCUSSION Pending published evidence from RCTs, this SRMA focused on available real-world observational studies, revealing a beneficial effect of TCZ use in preventing mortality in hospitalized adults with COVID-19. However, the present results also showed a higher relative risk for ICU admission and the occurrence of secondary infections in such COVID-19 patients receiving TCZ. To date, two existing SRMA have summarized current evidence on the beneficial and harmful effects of TCZ in COVID-19. The first by Lan SH Zhang et al., included 7 studies, with no conclusive evidence that TCZ would provide any additional benefit to patients with severe COVID-19 [77]. The second, registered in the medRxiv repository by Boregowda et al., included 16 studies, concluding that the addition of TCZ to SOC might reduce mortality in COVID-19 patients requiring hospitalization [78]. The present SRMA updated and expanded the revision to 57 observational studies. As expected, most included studies emerged recently and were performed in hospitals from high-income European countries and the US. The finding that most of the included patients were men in their 6-8th decades of life was consistent with what has already been described in the general population requiring hospital admission due to COVID-19 [79]. The vast majority of included studies used a single dose of 400-800 mg intravenous or 162-324 mg subcutaneous, and a few of them allowed a second or even a third dose in case of worsening. Interestingly, the present SRMA showed that TCZ was mainly prescribed as a second step to treat those patients at risk of transition to a more severe condition, after showing poor response to antiviral agents. Accordingly, in a notable proportion of included studies, TCZ was indicated in combination with corticosteroids. In this respect, in view of the RECOVERY trial [5] which reported significant benefit from the use of steroids in severe COVID-19, it is difficult to distinguish in depth the contribution of TCZ on the outcomes in such included patients receiving TCZ and steroids. On the other hand, in addition to TCZ, a very small proportion (<1%) of included patients from more recent studies also received remdesivir, as a proven antiviral against SARS-Cov-2. In addition to concomitant drugs, other relevant factors should be taken into account when considering the impact of TCZ use on clinical outcomes in COVID-19. In this respect, in addition to the age and underlying comorbidities of included patients, one of the most important factors to be considered at the clinical level is the severity of the clinical-stage when indicating TCZ to treat the COVID-19 illness [6,79]. Unfortunately, the severity of patients could not be accurately inferred from the clinical, laboratory, or radiological parameters documented in the included studies of the present SRMA. Therefore, mortality was compared between those patients admitted to hospital wards and ICUs, in those studies in which the hospital site from where TCZ was administered was specified. Logically, patients in whom TCZ was indicated during ICU admission showed higher mortality in comparison to those treated in hospital wards. However, since the COVID-19 pandemic induced an unprecedented influx of patients into the ICUs, the particular emergency and resource availability of each hospital involved in the included studies most likely conditioned either the criteria when transferring patients from hospital wards to ICUs, or the ethical decisions related to the withdrawal of life support decisions. Outside of RCTs, observational data from the included studies only reflected the clinical practice of physicians when indicating TCZ use. Consequently, the present results show that TCZ was mostly prescribed in those more seriously-ill patients presenting at a more advanced stage of COVID-19, with severe lung injury and systemic hyperinflammatory multi-organ failure. In this regard, it would be unfair to infer that the higher the use of TCZ and the average of concomitant corticosteroids, the higher the risk of ICU admission or death. Therefore, further RCTs on TCZ use in COVID-19 should clarify the interaction of confounding variables, it being crucial to know which patients are the best candidates to eventually receive TCZ as an immunomodulatory agent, as well as the beneficial and harmful effects of its use in the absence of or in combination with steroids. Moreover, in the particular case of the ICU setting, most of the included studies showed insufficient data to appropriately assess the effects when TCZ was prescribed in critically-ill patients. However, since a majority of such seriously-ill patients with COVID-19 admitted to ICUs are submitted to mechanical ventilation and other multiple invasive procedures, and receive concomitant wide-spectrum antibiotics or steroid treatment, it is of great concern the well-known risk of TCZ favoring the occurrence of life-threatening secondary bacterial, viral or fungal opportunistic infections, as it has also been documented in the present SRMA in up to one-fifth of cases in the TCZ-group [53,80,81]. As mentioned, the first and major limitation of this SRMA is the lack of data from RCTs. In their absence, the present revision was based on observational studies; therefore, conclusions should be considered with caution. Moreover, a second limitation is the fact that most of the included studies were retrospective in nature. A third limitation is the heterogeneity regarding the study population (I2 index) and the potential risk of detected bias. Fourth, variations in criteria for prescribing TCZ may not be ruled out in the included studies, although most of them indicated TCZ use to treat those patients with severe COVID-19 with the systemic hyperinflammatory state. Fifth, important factors influencing the effect of TCZ on clinical outcomes such as the baseline characteristics of the patients included, the average time from symptoms onset to TCZ administration, the clinical severity of the disease at the time of TCZ administration, the doses and the form of administration used, the hospital site from where TCZ was indicated, or the use of concomitant drug regimens could not be evaluated in-depth, since they were not uniformly provided by the included studies. Sixth, in the vast majority of included studies, there is a lack of subgroup analyses according to age, sex or underlying conditions, concomitant treatments, the requirement of mechanical ventilation or ICU admission, and comparisons between ventilated and non-ventilated patients. Finally, there is a wide range in the median time of follow-up after TCZ administration, which hinders assessment of consistent improvement, late-onset adverse events, and real in-hospital mortality in those patients with prolonged evolution. Thus, some patients considered “survivors” in some included studies may have ended up dying. In conclusion, pending evidence from RCTs, this systematic review provides updated and extended data from observational studies on the use of TCZ in COVID-19. The present results showed TCZ to be beneficial in reducing overall in-hospital mortality in adults with COVID-19, with an NNT to save one life of 20. These findings were more apparent in those non-critically-ill COVID-19 patients admitted to hospital wards, receiving TCZ at the early stage of hyperinflammatory response syndrome. By contrast, the TCZ-group was at higher risk for secondary infections, especially in those patients admitted to ICU. Notwithstanding these results, conclusions should be considered as weak evidence since they are based on observational studies, most of them retrospective. However, these findings may help physicians and researchers to optimize strategies towards precision medicine when designing further RCTs focused on the use of TCZ in COVID-19. ## Data Availability The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request. ## ACKNOWLEDGMENTS We dedicate this work to the memory of those patients worldwide who have not survived COVID-19. We thank CERCA Programme/Generalitat de Catalunya for institutional support. ## FUNDING SOURCE No funding or sponsorship was received for this study or publication of this article. ## DECLARATION OF INTERESTS Jordi Rello has received consultancy honoraria from Roche. The rest of the authors declare no competing interests. ## AUTHORS’ CONTRIBUTION Study conception and design: M.R-R., X.C. Acquisition, analyses and interpretation of data: M.R-R., J.M.M-L, A.M., N.A.H., J.R., X.C. Manuscript draft and critical revision: M.R-R., J.R., X.C. ## Footnotes * jmora{at}bellvitgehospital.cat (J.M. Mora-Lujan), asaez{at}bellvitgehospital.cat (A. Montero), nhoms{at}bellvitgehospital.cat (N.A. Homs), jrello{at}crips.es (J. Rello), xcorbella{at}bellvitgehospital.cat (X. Corbella) * Figures 3 and 4 were added * Received September 5, 2020. * Revision received September 25, 2020. * Accepted September 26, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## REFERENCES 1. [1].Azoulay E, de Waele J, Ferrer R, Staudinger T, Borkowska M, Povoa P et al. International variation in the management of severe COVID-19 patients. Crit Care 2020;24(1):486. [https://doi.org/10.1186/s13054-020-03194-w](https://doi.org/10.1186/s13054-020-03194-w). Accessed 12 August 2020. 2. [2].Gautret P, Cagier JC, Parola P, Hoang VT, Meddeb L, Mailhe M et al. Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial. Int J Antimicrob Agents 2020:105949. [https://doi.org/10.1016/j.ijantimicag.2020.105949](https://doi.org/10.1016/j.ijantimicag.2020.105949). Accessed 25 March 2020. 3. [3].Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G, et al. A Trial of Lopinavir-Ritonavir in adults hospitalized with severe Covid-19. N Engl J Med 2020; 382:1787–99. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 4. [4].Grein J, Ohmagari N, Shin D, Diaz G, Asperges E, Castagna A, et al. Compassionate Use of Remdesivir for Patients with Severe Covid-19. N Engl J Med. 2020;382:2327–36. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NE-JMoa2007016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 5. [5].Horby P, Lim WS, Emberson JR, Mafham M, Bell J, Linsell L, et al. Dexamethasone in hospitalized patients with Covid-19. Preliminary report. N Engl J Med 2020. [https://doi.org/10.1056/NEJMoa2021436](https://doi.org/10.1056/NEJMoa2021436). Accessed 25 August 2020 6. [6].Siddiqi HK, Mehra MR. COVID-19 illness in native and immunosuppressed states: A clinical-therapeutic staging proposal. J Heart Lung Transplant 2020;39(5):405–7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.healun.2020.03.012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 7. [7].Azoulay E, Darmon M, Valade S.- Acute life-threatening toxicity from CAR T-cell therapy. Intensive Care Med 2020. [https://doi.org/10.1007/s00134-020-06193-1](https://doi.org/10.1007/s00134-020-06193-1). Accessed 25 August 2020 8. [8].Jamilloux Y, Henry T, Belot A, Viel S, Fauter M, El Jammalet T, et al. Should we stimulate or suppress immune responses in COVID-19? Cytokine and anti-cytokine interventions. Autoimmun Rev 2020;19:102567. [https://doi.org/10.1016/j.autrev.2020.102567](https://doi.org/10.1016/j.autrev.2020.102567). Accessed 25 August 2020 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32376392&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 9. [9].Aziz M, Fatima R, Assaly R. Elevated Interleukin-6 and Severe COVID-19: A Meta-Analysis. J Med Virol. 2020. [https://doi.org/10.1002/jmv.25948](https://doi.org/10.1002/jmv.25948). Accessed 21 August 2020. 10. [10].Zhang C, Wu Z, Li JW, Zhao H, Wang GQ. The cytokine release syndrome (CRS) of severe COVID-19 and Interleukin-6 receptor (IL-6R) antagonist Tocilizumab may be the key to reduce the mortality. Int J Antimicrob Agents 2020;55:105954. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ijantimicag.2020.105954&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 11. [11].Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al. Effective treatment of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci USA 2020;117:10970–5. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTE3LzIwLzEwOTcwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMDkvMjYvMjAyMC4wOS4wNS4yMDE4ODkxMi5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 12. [12].Luo P, Liu Y, Qiu L, Liu X, Liu D, Li J. Tocilizumab treatment in COVID-19: a single center experience. J Med Virol 2020;92:814–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/jmv.25801&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 13. [13].Antony SJ, Davis MA, Davis MG, Almaghlouth NK, Guevara R, Omar F, et al. Early use of tocilizumab in the prevention of adult respiratory failure in SARS-CoV-2 infections and the utilization of interleukin - 6 levels in the management. IDCases. 2020; 21: e00888. [https://doi.Org/10.1016/j.idcr.2020.e00888](https://doi.Org/10.1016/j.idcr.2020.e00888). Accessed 25 August 2020 14. [14].Quartuccio L, Sonaglia A, Pecori D, Peghin M, Fabris M, Tascini C, et al. Higher levels of IL-6 early after tocilizumab distinguish survivors from non-survivors in COVID-19 pneumonia: a possible indication for deeper targeting IL-6. J Med Virol. 2020:10.1002/jmv.26149. [https://doi.org/10.1002/jmv.26149](https://doi.org/10.1002/jmv.26149). Accessed 25 August 2020 15. [15].Toniati P, Piva S, Cattalini M, Garrafa E, Regola F, Castelli F, et al. Tocilizumab for the treatment of severe COVID-19 pneumonia with hyperinflammatory syndrome and acute respiratory failure: a single center study of 100 patients in Brescia, Italy. Autoimmun Rev 2020; 19:102568. Accessed 25 August 2020 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.autrev.2020.102568&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 16. [16].Alattar R, Ibrahim TBH, Shaar SH, Abdalla S, Shukri K, Daghfalt JN, et al. Tocilizumab for the treatment of severe COVID-19. J Med Virol 2020: 10.1002/jmv.25964. [https://doi.org/10.1002/jmv.25964](https://doi.org/10.1002/jmv.25964). Accessed 25 August 2020 17. [17].Sciascia S, Aprà F, Baffa A, Baldovino S, Boaro D, Boero R, et al. Pilot prospective open, single-arm multicentre study on off-label use of tocilizumab in patients with severe COVID-19. Clin Exp Rheumatol 2020;38:529–32. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 18. [18].Morrison AR, Johnson JM, Griebe KM, Jones MC, Stine JJ, Hencken LN, et al. Clinical characteristics and predictors of survival in adults with coronavirus disease 2019 receiving tocilizumab. J Autoimmun 2020:102512. [https://doi.org/10.1016/j.jaut.2020.102512](https://doi.org/10.1016/j.jaut.2020.102512). Accessed 25 August 2020 19. [19].Morena V, Milazzo L, Oreni L, Bestetti G, Fossali T, Bassoli C, et al. Off-label use of tocilizumab for the treatment of SARS-CoV-2 pneumonia in Milan, Italy. Eur J Intern Med 2020;76:36–42. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 20. [20].Marfella R, Paolisso P, Sardu C, Bergamaschib L, D’Angelob EC, Barbieri M, et al. Negative impact of hyperglycaemia on tocilizumab therapy in Covid-19 patients. Diabetes Metab 2020: S1262-3636(20)30082-3. [https://doi.org/10.1016/j.diabet.2020.05.005](https://doi.org/10.1016/j.diabet.2020.05.005). Accessed 25 August 2020 21. [21].Sanchez-Montalvà A, Selares-Nadal J, Espinosa-Pereiro J, Fernandez-Hidalgo N, Perez-Hoyos S, Salvador F, et al. Early outcomes of tocilizumab in adults hospitalized with severe COVID19. An initial report from the Vall d’Hebron COVID19 prospective cohort study. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.05.07.20094599v2](https://www.medrxiv.org/content/10.1101/2020.05.07.20094599v2). Accessed 31 July 2020 22. [22].Jordan SC, Zakowski P, Tran HP, Smith EA, Gaultier C, Marks G, et al. Compassionate Use of Tocilizumab for Treatment of SARS-CoV-2 Pneumonia. Clin Infect Dis 2020: ciaa812. [https://doi.org/10.1093/cid/ciaa812](https://doi.org/10.1093/cid/ciaa812). Accessed 25 August 2020 23. [23].Nasir N, Syed Faisal M, Kiren H, Khanum I, Jamil B. Treatment of ARDS and hyperinflammation in COVID-19 with IL-6 antagonist Tocilizumab: a tertiary care experience from Pakistan. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.23.20134072v1](https://www.medrxiv.org/content/10.1101/2020.06.23.20134072v1) Accessed 25 August 2020 24. [24].Borku Uysal B, Ikitimur H, Yavuzer S, Ikitimur B, Uysal H, Islamoglu MS, et al. Tocilizumab challenge: A series of cytokine storm therapy experiences in hospitalized COVID-19 pneumonia patients. J Med Virol 2020 Jun 2:10.1002/jmv.26111. [https://doi.org/10.1002/jmv.26111](https://doi.org/10.1002/jmv.26111). Accessed 25 August 2020 25. [25].Issa N, Dumery M, Guisset O, Mourissoux G, Bonnet F, Camou F. Feasibility of Tocilizumab in ICU patients with COVID-19. J Med Virol 2020:10.1002/jmv.26110. [https://doi.org/10.1002/jmv.26110](https://doi.org/10.1002/jmv.26110). Accessed 25 August 2020 26. [26].Campins L, Boixeda R, Perez-Cordon L, Aranega R, Lopera C, Force L. Early tocilizumab treatment could improve survival among COVID-19 patients. Clin Exp Rheumatol 2020;38:578. Accessed 25 August 2020 27. [27].Sanz Herrero F, Puchades Gimeno F, Ortega García P, Ferrer Gómez C, Ocete Mochón MD, García Deltoro M. Methylprednisolone added to tocilizumab reduces mortality in SARS-CoV-2 pneumonia: An observational study. J Intern Med 2020; 10.1111/joim.13145. [https://doi.org/10.1111/joim.13145](https://doi.org/10.1111/joim.13145). Accessed 25 August 2020 28. [28].Knorr JP, Colomy V, Mauriello CM, Ha S. Tocilizumab in patients with severe COVID-19: a single-center observational analysis. J Med Virol. 2020 Jun 17:10.1002/jmv.26191. [https://doi.org/10.1002/jmv.26191](https://doi.org/10.1002/jmv.26191). Accessed 25 August 2020 29. [29].Lohse A, Klopfenstein T, Balblanc JC, Royer PY, Bossert M, Gendrin V, et al. Predictive factors of mortality in patients treated with tocilizumab for acute respiratory distress syndrome related to coronavirus disease 2019 (COVID-19). Microbes Infect 2020: S1286-4579(20)30123-4. [https://doi.org/10.1016/j.micinf.2020.06.005](https://doi.org/10.1016/j.micinf.2020.06.005). Accessed 25 August 2020 30. [30].Conrozier T, Lohse A, Balblanc JC, Dussert P, Royer P, Bossert M, et al. Biomarker variation in patients successfully treated with tocilizumab for severe coronavirus disease 2019 (COVID-19): results of a multidisciplinary collaboration. Clin Exp Rheumatol 2020; 38:742–7. 31. [31].Petrak R, Skorodin N, Van Hise N, Fliegelman R, Pinsky J, Didwania V, et al. Tocilizumab as a Therapeutic Agent for Critically Ill Patients Infected with SARS-CoV-2. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.05.20122622v1](https://www.medrxiv.org/content/10.1101/2020.06.05.20122622v1) Accessed 25 August 2020 32. [32].Górgolas M, Cabello A, Prieto Perez L, Villar Alvarez F, Alvarez Alvarez B, Rodriguez Nieto MJ, et al. Compassionate Use of Tocilizumab in Severe SARS-CoV2 Pneumonia. When late administration is too late. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.13.20130088v1](https://www.medrxiv.org/content/10.1101/2020.06.13.20130088v1) Accessed 25 August 2020 33. [33].Jiménez-Britez G, Ruiz P, Soler X. Tocilizumab plus glucocorticoids in severe and critically COVID-19 patients. A single center experience. Med Clin 2020:S0025-7753(20)30441-3. [https://doi.org/10.1016/j.medcli.2020.07.001](https://doi.org/10.1016/j.medcli.2020.07.001). Accessed 25 August 2020 34. [34].Rimland CA, Morgan CE, Bell GJ, Kim MK, Hedrick T, Marx A, et al. Clinical characteristics and early outcomes in patients with COVID-19 treated with tocilizumab at a United States academic center. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.05.13.20100404v2](https://www.medrxiv.org/content/10.1101/2020.05.13.20100404v2) Accessed 25 August 2020 35. [35].Sinha P, Mostaghim A, Bielick CG, McLaughlin A, Hamer DH, Wetzler LM, et al. Early administration of Interleukin-6 inhibitors for patients with severe Covid-19 disease is associated with decreased intubation, reduced mortality, and increased discharge. Int J Infect Dis 2020:S1201-9712(20)30568-3. [https://doi.org/10.1016/j.ijid.2020.07.023](https://doi.org/10.1016/j.ijid.2020.07.023). Accessed 25 August 2020 36. [36].Patel A, Shah K, Dharsandiya M, Patel K, Patel T, Patel M, et al. Safety and efficacy of tocilizumab in the treatment of severe acute respiratory syndrome coronavirus-2 pneumonia: A retrospective cohort study. Indian J Med Microbiol 2020; 38:117–23. [CrossRef](http://medrxiv.org/lookup/external-ref?access\_num=10.4103/ijmm.IJMM_20_298&link_type=DOI) 37. [37].Hashimoto S, Kitajima H, Arai T, Tamura Y, Nagai T, Morishita H, et al. A retrospective study evaluating efficacy and safety of compassionate use of tocilizumab in 13 patients with severe-to-critically ill COVID-19: analysis of well-responding cases and rapidly-worsening cases after tocilizumab administration. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.24.20134288v1](https://www.medrxiv.org/content/10.1101/2020.06.24.20134288v1) Accessed 25 August 2020 38. [38].Formina DS, Lysenko MA, Beloglazova IP, Mutinova ZY, Poteshkina NG, Samsonova IV, et al. Temporal clinical and laboratory response to interleukin-6 receptor blockade with Tocilizumab in 89 hospitalized patients with COVID-19 pneumonia. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.12.20122374v1](https://www.medrxiv.org/content/10.1101/2020.06.12.20122374v1) Accessed 25 August 2020 39. [39].Rubio-Rivas M, Ronda M, Padulles A, Riera-Mestre A, Garcia-Forero C, Iriarte A, et al. Beneficial Effect of Corticosteroids in Preventing Mortality in Patients Receiving Tocilizumab to Treat Severe COVID-19 Illness. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.08.31.20182428v1](https://www.medrxiv.org/content/10.1101/2020.08.31.20182428v1) Accessed 25 August 2020 40. [40].Rodríguez-Baño J, Pachón J, Carratalà J, Ryan P, Jarrín I, Yllescas M, et al. Treatment with tocilizumab or corticosteroids for COVID-19 patients with hyperinflammatory state: a multicentre cohort study (SAM-COVID-19). Clin MIcrobiol Infect 2020 Aug 26:S1198-743X(20)30492-4. [https://doi.org/10.1016/ì.cmi.2020.08.010](https://doi.org/10.1016/%C3%AC.cmi.2020.08.010). Accessed 25 August 2020 41. [41].Quartuccio L, Sonaglia A, Mcgonagle D, Fabris M, Peghin M, Pecori D, et al. Profiling COVID-19 pneumonia progressing into the cytokine storm syndrome: Results from a single Italian Centre study on tocilizumab versus standard of care. J Clin Virol 2020;129:104444. [https://doi.org/10.1016/Mcv.2020.104444](https://doi.org/10.1016/Mcv.2020.104444). Accessed 25 August 2020 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jcv.2020.104444&link_type=DOI) 42. [42].Martínez-Sanz J, Muriel A, Ron R, Herrera S, Ron R, Perez-Molina JA, et al. Effects of Tocilizumab on Mortality in Hospitalized Patients with COVID-19: A Multicenter Cohort Study. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.08.20125245v1](https://www.medrxiv.org/content/10.1101/2020.06.08.20125245v1) Accessed 31 August 2020 43. [43].Moreno-García E., Rico V, Albiach L, Aguero D, Ambrosioni J, Bodro M, et al. Tocilizumab is associated with reduced risk of ICU admission and mortality in 1 patients with SARS-CoV-2 infection. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.05.20113738v1](https://www.medrxiv.org/content/10.1101/2020.06.05.20113738v1) Accessed 31 August 2020 44. [44].Guaraldi G, Meschiari M, Cozzi-lepri A, Milic J, Tonelli R, Menozzi M, et al. Tocilizumab in patients with severe COVID-19: a retrospective cohort study. The Lancet Rheumatology 2020;9913(20):1–11. 45. [45].Campochiaro C, Della-Torre E, Cavalli G, De Luca G, Ripa M, Boffini N, et al. Efficacy and safety of tocilizumab in severe COVID-19 patients: a single-center retrospective cohort study. Eur J Intern Med 2020;76:43–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ejim.2020.05.021&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 46. [46].Mikulska M, Nicolini LA, Signori A, Di Biagio A, Sepulcri C, Russo C, et al. Tocilizumab and steroid treatment in patients with COVID-19 pneumonia. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.22.20133413v1](https://www.medrxiv.org/content/10.1101/2020.06.22.20133413v1). Accessed 31 August 2020 47. [47].Colaneri M, Bogliolo L, Valsecchi P, Sacchi P, Zuccaro V, Brandolino F, et al; COVID IRCCS San Matteo Pavia Task Force. Tocilizumab for treatment of severe COVID-19 patients: preliminary results from SMAtteo COvid19 REgistry (SMACORE). Microorganisms 2020; 8: 695. 48. [48].Price CC, Altice FL, Shyr Y, Koff A, Pischel L, Goshua G, et al. Tocilizumab Treatment for Cytokine Release Syndrome in Hospitalized COVID-19 Patients: Survival and Clinical Outcomes. Chest 2020:S0012-3692(20)31670-6. 49. [49].Maeda T, Obata R, Rizk D, Kuno T. The Association of Interleukin-6 value, Interleukin inhibitors and Outcomes of Patients with COVID-19 in New York City. J Med Virol 2020; [https://doi.org/10.1002/jmv.26365](https://doi.org/10.1002/jmv.26365) Accessed 31 August 2020 50. [50].Capra R, De Rossi N, Mattioli F, Romanelli G, Scarpazza C, Sormani MP, et al. Impact of low dose tocilizumab on mortality rate in patients with COVID-19 related pneumonia. Eur J Intern Med 2020;76:31–5. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 51. [51].Rossotti R, Travi G, Ughi N, Corradin M, Baiguera C, Fumagalli R, et al. Safety and efficacy of anti-il6-receptor tocilizumab use in severe and critical patients affected by coronavirus disease 2019: a comparative analysis. J Infect. 2020:S0163-4453(20)30467-9. [https://doi.org/10.1016/j.jinf.2020.07.008](https://doi.org/10.1016/j.jinf.2020.07.008). Accessed 31 August 2020 52. [52].Eimer J, Vesterbacka J, Svensson AK, Stojanovic B, Wagrell C, Sönnerborg A, et al. Tocilizumab shortens time on mechanical ventilation and length of hospital stay in patients with severe COVID-19: a retrospective cohort study. J Int Med 2020; [https://doi.org/10.1111/joim.13162](https://doi.org/10.1111/joim.13162) Accessed 31 August 2020 53. [53].Kimmig LM, Wu D, Gold M, Pettit NN, Pitrak D, Mueller J, et al. IL6 inhibition in critically ill COVID-19 patients is associated with increased secondaryInfections. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.05.15.20103531v2](https://www.medrxiv.org/content/10.1101/2020.05.15.20103531v2) Accessed 31 August 2020 54. [54].Somers EC, Eschenauer GA, Troost JP, Golob JL, Gandhi TN, Wang Lu, et al. Tocilizumab for treatment of mechanically ventilated patients with COVID-19. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.05.29.20117358v1](https://www.medrxiv.org/content/10.1101/2020.05.29.20117358v1) Accessed 25 August 2020 55. [55].Tsai A, Diawara O, Nahass RG, Brunetti L. Impact of tocilizumab administration on mortality in severe COVID-19. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.07.30.20114959v1](https://www.medrxiv.org/content/10.1101/2020.07.30.20114959v1) Accessed 31 August 2020 56. [56].Kewan T, Covut F, Al-Jaghbeer MJ, Rose L, Gopalakrishna KV, Akbik B. Tocilizumab for treatment of patients with severe COVID19: A retrospective cohort study. EClinicalMedicine 24 (2020) 100418 57. [57].Patel K, Gooley TA, Bailey N, Bailey M, Hegerova L, Batchelder A, et al. Use of the IL-6R Antagonist Tocilizumab in Hospitalized COVID-19 Patients. J Inter Med 03 Aug 2020, [https://doi.org/10.1111/joim.13163](https://doi.org/10.1111/joim.13163) Accessed 31 August 2020 58. [58].Wadud N, Ahmed N, Shergil M, Khan M, Krishna MG, Gilani A, et al. Improved survival outcome in SARs-CoV-2 (COVID-19) Acute Respiratory Distress Syndrome patients with Tocilizumab administration. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.05.13.20100081v1](https://www.medrxiv.org/content/10.1101/2020.05.13.20100081v1) Accessed 31 August 2020 59. [59].Ramaswamy M, Mannam P, Comer R, Sinclair E, McQuaid DB, Schmidt ML, et al. Off-Label Real World Experience Using Tocilizumab for Patients Hospitalized with COVID-19 Disease in a Regional Community Health System: A Case-Control Study. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.05.14.20099234v1](https://www.medrxiv.org/content/10.1101/2020.05.14.20099234v1) Accessed 31 August 2020 60. [60].Rojas-Marte GR, Khalid M, Mukhtar O, Hashmi AT, Waheed MA, Ehrlich S,et al. Outcomes in Patients with Severe COVID-19 Disease Treated with Tocilizumab - A Case-Controlled Study. QJM. 2020: hcaa206. [https://doi.org/10.1093/qjmed/hcaa206](https://doi.org/10.1093/qjmed/hcaa206) Accessed 31 August 2020 61. [61].Roumier M, Paule R, Groh M, Vallee A, Ackermann F. Interleukin-6 blockade for severe COVID-19. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.04.20.20061861v1](https://www.medrxiv.org/content/10.1101/2020.04.20.20061861v1) Accessed 31 August 2020 62. [62].Rossi B, Nguyen LS, Zimmermann P, Boucenna F, Baucher L, Dubret L, et al. Effect of tocilizumab in hospitalized patients with severe pneumonia COVID-19: a cohort study. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.06.06.20122341v1](https://www.medrxiv.org/content/10.1101/2020.06.06.20122341v1) Accessed 31 August 2020 63. [63].Potere N, Di Nisio M, Cibelli D, Scurti R, Frattari A, Porreca E, et al. Interleukin-6 receptor blockade with subcutaneous tocilizumab in severe COVID-19 pneumonia and hyperinflammation: a case-control study. Ann Rheum Dis. 2020:218243. [https://doi.org/10.1136/annrheumdis-2020-218243](https://doi.org/10.1136/annrheumdis-2020-218243). Accessed 31 August 2020 64. [64].Klopfenstein T, Zayet S, Lohse A, Balblanc JC, Badie J, Royer PY, et al. Tocilizumab 300 therapy reduced intensive care unit admissions and/or mortality in COVID-19 patients. Med Mal Infect 2020:S0399-077X(20)30129-3. 65. [65].Canziani LM, Trovati S, Brunetta E, De Santis M, Bombardieri E, Guidelli G, et al. Interleukin-6 receptor blocking with intravenous tocilizumab in COVID-19 severe acute respiratory distress syndrome: A retrospective case-control survival analysis of 128 patients. J Autoimmun 2020 Jul 8;102511. [https://doi.org/10.1016/j.jaut.2020.102511](https://doi.org/10.1016/j.jaut.2020.102511) Accessed 31 August 2020 66. [66].Ip A, Berry DA, Hansen E, Goy AH, Pecora AL, Sinclaire BA, et al. Hydroxychloroquine and Tocilizumab Therapy in COVID-19 Patients - An Observational Study. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.05.21.20109207v1](https://www.medrxiv.org/content/10.1101/2020.05.21.20109207v1) Accessed 31 August 2020 67. [67].Carvalho V, Turon R, Gonçalves B, Ceotto V, Kurtz P, Righy C. Effects of Tocilizumab in Critically Ill Patients With COVID-19: A Quasi-Experimental Study. medRxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.07.13.20149328v1](https://www.medrxiv.org/content/10.1101/2020.07.13.20149328v1) Accessed 31 August 2020 68. [68].COVID-19 Treatment Guidelines Panel. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines. National Institutes of Health. Available at [https://www.covid19treatmentguidelines.nih.gov/](https://www.covid19treatmentguidelines.nih.gov/). Accessed 25 August 2020. 69. [69].Roche. Roche provides an update on the Phase III COVACTA trial of Actemra/RoActemra in hospitalized patients with severe COVID-19 associated pneumonia. 2020. Available at: [https://www.roche.com/investors/updates/inv-update-2020-07-29.htm](https://www.roche.com/investors/updates/inv-update-2020-07-29.htm). Accessed 10 August 2020. 70. [70].Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ 2009 Jul 21;339:b2700. [https://doi.org/10.1136/bmj.b2700](https://doi.org/10.1136/bmj.b2700) Accessed 30 July 2020 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjE3OiIzMzkvanVsMjFfMS9iMjcwMCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA5LzI2LzIwMjAuMDkuMDUuMjAxODg5MTIuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 71. [71].Wells G, Shea B, O’Connell D. The Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomized studies in meta-analysis. Available at [http://www.ohri.ca/programs/clinicalepidemiology/oxford.asp](http://www.ohri.ca/programs/clinicalepidemiology/oxford.asp). Accessed 29 August 2020 72. [72].Wilson EB. *Probable inference, the law of* succession, and statistical inference. J Am Stat Assoc 1927;22:209–12. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2307/2276774&link_type=DOI) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000188338900021&link_type=ISI) 73. [73].Garwood F. Fiducial Limits for the Poisson Distribution. Biometrika 1936,28:437-42. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/biomet/28.3-4.437&link_type=DOI) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000200863800007&link_type=ISI) 74. [74].Kahn HA, Sempos CT. Statistical Q14 methods in epidemiology. Oxford University Press; 1989. 75. [75].Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327:557–60. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiYm1qIjtzOjU6InJlc2lkIjtzOjEyOiIzMjcvNzQxNC81NTciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8wOS8yNi8yMDIwLjA5LjA1LjIwMTg4OTEyLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 76. [76].Begg CB, Berlin JA. Publication bias and dissemination of clinical research. J Natl Cancer Inst 1989;81:107–15. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/jnci/81.2.107&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2642556&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1989R731800003&link_type=ISI) 77. [77].Lan SH, Lai CC, Huang HT, Chang SP, Lu LC, Hsuehft PR. Tocilizumab for severe COVID-19: a systematic review and metaanalysis. International Journal of Antimicrobial Agents, [https://doi.org/10.1016/j.ijantimicag.2020.106103](https://doi.org/10.1016/j.ijantimicag.2020.106103) Accessed 30 July 2020 78. [78].Boregowda U, Perisetti A, Nanjappa A, Gajendran M, Goyal H. Addition of Tocilizumab to the standard of care reduces mortality in severe COVID-19: A systematic review and meta-analysis. medrxiv preprint at [https://www.medrxiv.org/content/10.1101/2020.07.10.20150680v3](https://www.medrxiv.org/content/10.1101/2020.07.10.20150680v3) Accessed 30 July 2020 79. [79].Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 2020;395(10229):1054–62. [https://doi.org/10.1016/S0140-6736(20)30566-3](https://doi.org/10.1016/S0140-6736(20)30566-3). Accessed 30 July 2020 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(20)30566-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 80. [80].Antinori S, Bonazzetti C, Gubertini G, Capetti A, Pagani C, Morena V, et al. Tocilizumab for cytokine storm syndrome in COVID-19 pneumonia: an increased risk for candidemia? Autoimmun Rev. 2020;19:102564. [https://doi.org/10.1016/j.autrev.2020.102564](https://doi.org/10.1016/j.autrev.2020.102564). Accessed 30 July 2020 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F09%2F26%2F2020.09.05.20188912.atom) 81. [81].Han W, Quan B, Guo Y, Zhang J, Lu Y, Feng G, et al. The course of clinical diagnosis and treatment of a case infected with coronavirus disease 2019. J Med Virol. 2020;92:461–3. [https://doi.org/10.1002/jmv.25711](https://doi.org/10.1002/jmv.25711) Accessed 31 August 2020