The coronavirus disease 2019 (COVID-19) has caused a global pandemic, resulting in considerable morbidity and mortality. Tocilizumab, an inhibitor of IL-6, has been widely repurposed as a treatment of severely ill patients without robust evidence supporting its use. In this study, we aimed to systematically describe the effectiveness of treatment and prevention of the cytokine storms in COVID-19 patients with tocilizumab. In this multicentered retrospective and observational cohort study, 65 patients with COVID-19 receiving tocilizumab and 130 not receiving tocilizumab were propensity score matched at a ratio of 2:1 based on age, sex, and comorbidities from January 20, 2020 to March 18, 2020 in Wuhan, China. After adjusting for confounding, the detected risk for in-hospital death was lower in the tocilizumab group versus nontocilizumab group (hazard ratio = 0.47; 95% confidence interval = 0.25–0.90; p = 0.023). Moreover, use of tocilizumab was associated with a lower risk of acute respiratory distress syndrome (odds ratio = 0.23; 95% confidence interval = 0.11–0.45; p < 0.0001). Furthermore, patients had heightened inflammation and more dysregulated immune cells before treatment, which might aggravate disease progression. After tocilizumab administration, abnormally elevated IL-6, C-reactive protein, fibrinogen, and activated partial thromboplastin time decreased. Tocilizumab may be of value in prolonging survival in patients with severe COVID-19, which provided a novel strategy for COVID-19–induced cytokine release syndrome. Our findings could inform bedside decisions until data from randomized, controlled clinical trials become available.

Coronavirus disease 2019 (COVID-19) has caused a global pandemic as a clinical syndrome caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (1). By August 18, 2020, the number of deaths had climbed to 767,158 among 21,549,706 confirmed cases in World Health Organization reports (2). According to a report, the mortality for critical cases reached 60.5% (3). The elevated inflammatory cytokines suggest that cytokine release syndrome (CRS) plays a major role in the pathology of COVID-19 (4, 5).

Although the current focus has been on the development of novel therapeutics, including antivirals and vaccines, there is still a long way to go before the vaccine is officially launched on the market. Before that, research is under way to repurpose medications. To dampen excessive serum inflammatory mediators, interest has increased in the use of anti-inflammatory agents. However, corticosteroids have short- and long-term adverse reactions (6), and plasmapheresis or continuous renal replacement therapy either requires specific equipment or lacks documented efficacy (7).

A better understanding of the underlying pathogenesis in CRS facilitates the design of immunotherapies. IL-6 is the key molecule of CRS, so IL-6R antagonist may be of value in improving outcomes (8, 9). Tocilizumab is a recombinant humanized monoclonal anti–IL‐6R Ab (10) that has been used worldwide in various rheumatic diseases and severe CAR T cell–induced CRS (11, 12). Given the pivotal role of IL-6 in COVID-19 and the efficacy of tocilizumab in CRS, tocilizumab was included for the first time in Diagnosis and Treatment Protocol for Novel Coronavirus Pneumonia (seventh edition) sponsored by National Health Commission of the People’s Republic of China. However, because of the small sample sizes of previous studies, there are insufficient data to support the efficacy of the tocilizumab in patients with COVID-19. To further provide a therapeutic strategy for this fatal disease, ultimately curbing the rising fatality rate of COVID-19, this multicenter, retrospective, observational study presents the clinical details of patients receiving tocilizumab from three hospitals in Wuhan, China.

This multicenter, retrospective, cohort study was conducted in three hospitals in Wuhan, China that were designated centers for COVID-19 treatment: Tongji Hospital, Wuhan Pulmonary Hospital, and Renmin Hospital of Wuhan University (the east campus). We used the following inclusion and exclusion criteria to determine the study cohort. The inclusion criteria included all adult patients (aged ≥18 y old, 5235 patients) with COVID-19, who were admitted to the above-mentioned hospitals in Hubei, China from January 20, 2020 to March 18, 2020. Eligibility criteria for tocilizumab administration were as follows: a diagnosis of COVID-19 confirmed upon RT-PCR positivity for SARS-CoV-2; patients with extensive lung lesions; severe cases who also show an increased level of IL-6 in laboratory testing. Exclusion criteria were as follows: incomplete medical records (e.g., transfer to any other hospital), evidence of concomitant bacterial infection, and pregnancy.

After quality control in clinical data, 65 patients using tocilizumab (B2084B21; Roche Pharma) were ultimately enrolled, and a total of 130 patients not taking tocilizumab were statistically matched by propensity score matching (13) at a ratio of 2:1, based on age, sex, and comorbidities. The comorbidities, including hypertension, diabetes, tumor, coronary heart disease, chronic obstructive pulmonary disease, cerebral infarction, liver cirrhosis, hepatitis, and tuberculosis, were used as matching factors in this study. These comorbidities have been reported to be risk factors for severity or death of COVID-19 (14, 15). The date of final follow-up was April 14, 2020.

For diagnosis, patients were assessed for eligibility on the basis of a positive RT-PCR assay (Shanghai Zhijian Biotechnology or Sansure Biotech) for SARS-CoV-2 in a respiratory tract sample tested by the local Center for Disease Control or by a designated diagnostic laboratory.

This study was approved by the Ethics Committee of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (TJ-C20200108) and granted a waiver of informed consent from study participants.

Epidemiological, clinical, radiological, laboratory, clinical treatments, and clinical outcomes data of all patients with laboratory-confirmed SARS-CoV-2 were obtained with data collection forms from electronic medical records of Tongji Hospital, Wuhan Pulmonary Hospital, and Renmin Hospital of Wuhan University (the east campus). The admission and in-hospital data of these patients were collected, reviewed, and verified by a trained team of physicians to guarantee the accuracy of the data extraction procedures. Any missing or uncertain records were collected and clarified through communication with involved health care providers and their families. The detailed and standardized information of demographic data, comorbidities, initial symptoms and vital signs were recorded or diagnosed at hospital admission. The chest computed tomography (CT) was recorded at hospital admission and after treatment. The complications, treatments, clinical outcomes, and hospital length of stay were monitored until April 14, 2020, the final data of follow-up. Laboratory examinations, including blood routine, immune cells subsets, inflammatory or infection-related cytokines and biomarkers, and cardiac/renal/liver/coagulation function tests, were detected on the last diagnosed date from laboratory information system.

The primary end point was in-hospital death. The main secondary outcomes were total days in the intensive care unit (ICU), the time from admission to discharge, and complications during hospitalization. Acute respiratory distress syndrome (ARDS) was defined according to the Berlin definition (16). Disseminated intravascular coagulation (DIC) was diagnosed by increased levels of platelets, clotting factors, and other blood components (17). Acute renal injury was identified according to the Kidney Disease: Improving Global Outcomes clinical practice guidelines (18). Acute liver damage was diagnosed by progressively elevated levels of alanine transaminase (ALT), aspartate aminotransferase (AST), or bilirubin. If the serum levels of cardiac biomarker high-sensitivity cardiac troponin I (hs-cTnI) were above the 99th percentile upper reference limit or new abnormalities were shown in electrocardiography and echocardiography, the acute cardiac injury was defined (19).

Safety outcomes included adverse events that occurred during treatment. Adverse events were classified according to the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0.

The onset of COVID-19 was defined as the time point when the symptoms were first noticed. Patients receiving tocilizumab during hospitalization were classified as the tocilizumab group. Patients who did not receive tocilizumab during hospitalization were classified as the nontocilizumab group. The criteria for discharge were an absence of fever for at least 3 d, substantial improvement in respiratory symptoms, and both lungs from chest CT and two throat-swab samples negative for SARS-CoV-2 RNA obtained at least 24 h apart (14). In this study, there were no cases lost to follow-up attributed to standardized government management and close tracking for the COVID-19 pandemic. The illness severity of COVID-19 was defined according to the criteria defined by the Diagnosis and Treatment Protocol for Novel Coronavirus Pneumonia (sixth interim edition) sponsored by National Health Commission of the People’s Republic of China.

Continuous variables were presented as median and interquartile range (IQR) or mean and SD. Categorical variables were expressed as a number (percentage). Differences between the tocilizumab group and the nontocilizumab group were compared. For continuous variables, a Student t test was used for normal distributed data and a Mann–Whitney U nonparametric test was used for nonnormal distributed continuous data. The Pearson χ2 test, Pearson χ2 test with Yates continuity correction, or Fisher exact test were applied for categorical variables. Time to events (discharge or death) were defined as the time from hospital admission to events. A Cox proportional hazards model was fitted for time to death, controlling for treatment group and potential confounders, including age, gender, and comorbidities. Univariable and multivariable logistic regression models were employed to estimate odds ratios (ORs) and 95% confidence intervals (CIs), adjusting for age, sex, and comorbidities. A two-sided p < 0.05 was considered statistically significant. All statistical analyses were performed using R (3.6.0) or SPSS Statistics (22.0).

This study cohort included 195 COVID-19 patients who were admitted to Tongji Hospital, Wuhan Pulmonary Hospital, and Renmin Hospital of Wuhan University (the east campus) during January 20, 2020 through March 18, 2020. Among these patients with COVID-19, 65 were classified as the tocilizumab group (71.0 y old [IQR 63.0–75.0]; 73.85% men), and 130 patients were classified as the nontocilizumab group (67.5 y old [IQR 61.0–75.0]; 63.08% men) according to the medication used during the hospitalization.

The characteristics of the tocilizumab group versus the nontocilizumab group on admission were provided in Supplemental Table I and Table I. In terms of age, gender, comorbidities, symptoms, and demographic characteristics (CT) of these patients, no significant differences were observed between these two groups. Compared with the nontocilizumab group, the tocilizumab group had higher level of D-dimer and urea. In the tocilizumab group, tocilizumab (B2084B21; Roche Pharma) was initiated at a median of 15 d (IQR 4.0–21.5) following admission. Tocilizumab was administered at a dose of 4–8 mg/kg body weight, and the recommended dose was 400 mg through an i.v. drip up to a maximum of 800 mg. Dilution was to 100 ml with 0.9% normal saline, and the infusion time was more than 1 h. In case of fever within 12 h, an additional dose was given, and the cumulative dose could not be given more than two times. A third infusion, 24 h apart from the second, was optional, based on clinical response. Additional information about dosing and administration appears in Supplemental Table II. In terms of other in-hospital treatment, there were no significant differences in both groups, suggesting that the different clinical outcomes were mainly triggered by the tocilizumab treatment (Supplemental Table III).

Table I.
Characteristics of the overall population at baseline
IndicatorsTotalTocilizumabNontocilizumabp Value
Characteristics on admission     
 Age (n = 195) in years 69.00 (62.00–75.00) 71.00 (63.00–75.00) 67.50 (61.00–75.00) 0.378 
 Male (n = 195) 130 (66.67%) 48 (73.85%) 82 (63.08%) 0.133 
 Female (n = 195) 65 (33.33%) 17 (26.15%) 48 (36.92%)  
 Systolic pressure (n = 193) in mm Hg 131.26 ± 18.15 134.40 ± 18.91 129.68 ± 17.62 0.087 
 Diastolic pressure (n = 193) in mm Hg 79.91 ± 12.09 82.74 ± 11.86 78.50 ± 12.00 0.096 
 Heart rate (n = 194) 93.00 (82.75–103.00) 90.00 (79.25–101.00) 94.50 (82.75–108.00) 0.075 
 Respiratory rate (n = 193) 21.00 (20.00–28.00) 22.00 (20.00–29.00) 21.00 (20.00–26.25) 0.758 
 Temperature (n = 191) in °C 36.80 (36.30–37.50) 36.70 (36.30–37.05) 36.80 (36.30–37.80) 0.136 
Laboratory examination on admission     
Inflammatory cytokines and biomarkers    
 IL-6 (n = 195) in pg/ml 41.02 (21.31–82.43) 31.95 (18.83–74.24) 44.18 (23.81–84.72) 0.091 
 IL-8 (n = 180) in pg/ml 20.15 (11.15–39.8) 18.85 (7.75–37.93) 20.25 (12.58–40.08) 0.195 
 IL-10 (n = 180) in pg/ml 7.10 (5.00–11.63) 6.35 (5.00–9.50) 7.65 (5.00–12.20) 0.082 
 IL-1β (n = 180) in pg/ml 7.88 ± 9.13 6.65 ± 4.61 8.36 ± 10.34 0.128 
 IL-2R (n = 179) in U/ml 835.00 (565.00–1234.0) 715.00 (468.5–1020.5) 887.50 (588.75–1331) 0.075 
 hs-CRP (n = 192) in mg/l 68.10 (34.58–133.60) 60.85 (23.35–139.23) 72.75 (38.63–124.10) 0.220 
 PCT (n = 171) in ng/ml 0.14 (0.06–0.41) 0.14 (0.07–0.36) 0.13 (0.06–0.42) 0.685 
Lymphocytes count per μl     
 Lymphocytes (n = 194) ×109/l 0.75 (0.55–1.06) 0.65 (0.47–1.06) 0.78 (0.61–1.05) 0.104 
 CD4+T cell (n = 64) (/μl) 243.00 (93.50–348.50) 264.00 (105.50–311.28) 217.00 (81.99–392.25) 0.731 
 CD8+T cell (n = 64) (/μl) 117.00 (43.27–282.00) 117.00 (56.27–255.50) 121.50 (37.75–308.25) 0.819 
 CD3+T cell (n = 64) (/μl) 423.00 (233.00–799.00) 379.00 (225.00–554.72) 438.50 (263.50–819.00) 0.366 
 CD3CD16+CD56+NK cell (n = 55) (/μl) 86.00 (38.00–143.00) 68.00 (44.00–127.00) 93.50 (27.00–150.25) 0.966 
 T cell+B cell+NK cells (n = 55) (/μl) 774.59 ± 478.54 663.92 ± 426.51 825.96 ± 499.78 0.294 
 CD19+B cells (n = 55) (/μl) 101.00 (53.00–163.00) 75.00 (59.00–118.00) 122.50 (49.50–164.50) 0.492 
Blood routine     
 Neutrophils (n = 194) (×109/l) 6.20 ± 4.26 6.57 ± 3.68 6.04 ± 4.50 0.388 
 Eosinophils (n = 194) (×109/l) 0.01 (0–0.04) 0.015 (0–0.055) 0.01 (0–0.03) 0.455 
 Basophils (n = 194) (×109/l) 0.01 (0.01–0.02) 0.01 (0.01–0.02) 0.01 (0.01–0.02) 0.774 
 Monocytes (n = 194) (×109/l) 0.43 (0.28–0.57) 0.41 (0.29–0.55) 0.45 (0.28–0.57) 0.345 
Organ damage indices     
 D-dimer (n = 179), μg/ml 1.61 (0.75–4.53) 2.57 (0.96–10.69) 1.38 (0.70–3.53) 0.046a 
 APTT (n = 180), s 42.00 (36.9–47.13) 41.20 (35.90–49.80) 42.50 (37.70–46.70) 0.881 
 PT (n = 181), s 14.50 (13.68–15.7) 14.30 (13.73–15.30) 14.50 (13.60–15.80) 0.456 
 Fbg (n = 179), g/l 5.38 ± 1.63 5.41 ± 1.74 5.36 ± 1.59 0.881 
 ALT (n = 195), U/l 27.00 (17.00–40.25) 30.00 (21.25–46.25) 24.00 (16.00–39.00) 0.078 
 AST (U/l, n = 195) 36.00 (25.00–53.25) 36.00 (26.00–54.75) 36.00 (25.00–51.25) 0.503 
 NT-proBNP (n = 163), pg/ml 368 (112.25–1013.75) 480.00 (145.00–888.00) 345.00 (99.00–1227.00) 0.930 
 CK (n = 142), U/l 108.50 (46.25–208.00) 80.50 (47.75–181.50) 123.00 (46.00–224.5) 0.523 
 CK-MB (n = 135), U/l 1.20 (0.63–2.48) 1.30 (0.85–2.30) 1.10 (0.60–2.50) 0.298 
 hs-cTnI (n = 163), pg/ml 10.35 (4.2–30.83) 8.00 (4.95–23.50) 11.40 (3.75–34.55) 0.868 
IndicatorsTotalTocilizumabNontocilizumabp Value
Characteristics on admission     
 Age (n = 195) in years 69.00 (62.00–75.00) 71.00 (63.00–75.00) 67.50 (61.00–75.00) 0.378 
 Male (n = 195) 130 (66.67%) 48 (73.85%) 82 (63.08%) 0.133 
 Female (n = 195) 65 (33.33%) 17 (26.15%) 48 (36.92%)  
 Systolic pressure (n = 193) in mm Hg 131.26 ± 18.15 134.40 ± 18.91 129.68 ± 17.62 0.087 
 Diastolic pressure (n = 193) in mm Hg 79.91 ± 12.09 82.74 ± 11.86 78.50 ± 12.00 0.096 
 Heart rate (n = 194) 93.00 (82.75–103.00) 90.00 (79.25–101.00) 94.50 (82.75–108.00) 0.075 
 Respiratory rate (n = 193) 21.00 (20.00–28.00) 22.00 (20.00–29.00) 21.00 (20.00–26.25) 0.758 
 Temperature (n = 191) in °C 36.80 (36.30–37.50) 36.70 (36.30–37.05) 36.80 (36.30–37.80) 0.136 
Laboratory examination on admission     
Inflammatory cytokines and biomarkers    
 IL-6 (n = 195) in pg/ml 41.02 (21.31–82.43) 31.95 (18.83–74.24) 44.18 (23.81–84.72) 0.091 
 IL-8 (n = 180) in pg/ml 20.15 (11.15–39.8) 18.85 (7.75–37.93) 20.25 (12.58–40.08) 0.195 
 IL-10 (n = 180) in pg/ml 7.10 (5.00–11.63) 6.35 (5.00–9.50) 7.65 (5.00–12.20) 0.082 
 IL-1β (n = 180) in pg/ml 7.88 ± 9.13 6.65 ± 4.61 8.36 ± 10.34 0.128 
 IL-2R (n = 179) in U/ml 835.00 (565.00–1234.0) 715.00 (468.5–1020.5) 887.50 (588.75–1331) 0.075 
 hs-CRP (n = 192) in mg/l 68.10 (34.58–133.60) 60.85 (23.35–139.23) 72.75 (38.63–124.10) 0.220 
 PCT (n = 171) in ng/ml 0.14 (0.06–0.41) 0.14 (0.07–0.36) 0.13 (0.06–0.42) 0.685 
Lymphocytes count per μl     
 Lymphocytes (n = 194) ×109/l 0.75 (0.55–1.06) 0.65 (0.47–1.06) 0.78 (0.61–1.05) 0.104 
 CD4+T cell (n = 64) (/μl) 243.00 (93.50–348.50) 264.00 (105.50–311.28) 217.00 (81.99–392.25) 0.731 
 CD8+T cell (n = 64) (/μl) 117.00 (43.27–282.00) 117.00 (56.27–255.50) 121.50 (37.75–308.25) 0.819 
 CD3+T cell (n = 64) (/μl) 423.00 (233.00–799.00) 379.00 (225.00–554.72) 438.50 (263.50–819.00) 0.366 
 CD3CD16+CD56+NK cell (n = 55) (/μl) 86.00 (38.00–143.00) 68.00 (44.00–127.00) 93.50 (27.00–150.25) 0.966 
 T cell+B cell+NK cells (n = 55) (/μl) 774.59 ± 478.54 663.92 ± 426.51 825.96 ± 499.78 0.294 
 CD19+B cells (n = 55) (/μl) 101.00 (53.00–163.00) 75.00 (59.00–118.00) 122.50 (49.50–164.50) 0.492 
Blood routine     
 Neutrophils (n = 194) (×109/l) 6.20 ± 4.26 6.57 ± 3.68 6.04 ± 4.50 0.388 
 Eosinophils (n = 194) (×109/l) 0.01 (0–0.04) 0.015 (0–0.055) 0.01 (0–0.03) 0.455 
 Basophils (n = 194) (×109/l) 0.01 (0.01–0.02) 0.01 (0.01–0.02) 0.01 (0.01–0.02) 0.774 
 Monocytes (n = 194) (×109/l) 0.43 (0.28–0.57) 0.41 (0.29–0.55) 0.45 (0.28–0.57) 0.345 
Organ damage indices     
 D-dimer (n = 179), μg/ml 1.61 (0.75–4.53) 2.57 (0.96–10.69) 1.38 (0.70–3.53) 0.046a 
 APTT (n = 180), s 42.00 (36.9–47.13) 41.20 (35.90–49.80) 42.50 (37.70–46.70) 0.881 
 PT (n = 181), s 14.50 (13.68–15.7) 14.30 (13.73–15.30) 14.50 (13.60–15.80) 0.456 
 Fbg (n = 179), g/l 5.38 ± 1.63 5.41 ± 1.74 5.36 ± 1.59 0.881 
 ALT (n = 195), U/l 27.00 (17.00–40.25) 30.00 (21.25–46.25) 24.00 (16.00–39.00) 0.078 
 AST (U/l, n = 195) 36.00 (25.00–53.25) 36.00 (26.00–54.75) 36.00 (25.00–51.25) 0.503 
 NT-proBNP (n = 163), pg/ml 368 (112.25–1013.75) 480.00 (145.00–888.00) 345.00 (99.00–1227.00) 0.930 
 CK (n = 142), U/l 108.50 (46.25–208.00) 80.50 (47.75–181.50) 123.00 (46.00–224.5) 0.523 
 CK-MB (n = 135), U/l 1.20 (0.63–2.48) 1.30 (0.85–2.30) 1.10 (0.60–2.50) 0.298 
 hs-cTnI (n = 163), pg/ml 10.35 (4.2–30.83) 8.00 (4.95–23.50) 11.40 (3.75–34.55) 0.868 

Sixty-five patients using tocilizumab and 130 patients not taking tocilizumab were compared. Epidemiological and laboratory data on admission of all patients with laboratory-confirmed SARS-CoV-2 were obtained with data collection forms from electronic medical records of Tongji Hospital, Wuhan Pulmonary Hospital, and Renmin Hospital of Wuhan University (the east campus). Continuous variables were described as median and IQR or mean and SD, and differences were assessed using analysis of Student t test or Mann–Whitney U test. Categorical variables were expressed as number (percentage), and differences between groups were assessed using Pearson χ2 test, Fisher exact test, or the continuity correction χ2 tests.

a

Data shown are significant at p < 0.05.

Abbreviations used in this table: CD, cluster of differentiation; CK, creatine kinase; CK-MB, creatine kinase–myocardial band; Fbg, fibrinogen; hs-CRP, hypersensitive CRP; NT-proBNP, N-terminal brain natriuretic propeptide; PCT, procalcitonin; PT, plasma prothrombin time.

During a follow-up duration, 56 died out of the 195 patients (28.72%) with COVID-19. The risk of in-hospital proportion of death was lower in tocilizumab group versus nontocilizumab group (21.54% [14/65] versus 32.31% [42/130]; p = 0.12) (Table II). In the Cox proportional hazard model, after adjusting for age, gender, and comorbidities, use of tocilizumab was associated with lower in-hospital proportion of death (hazard ratio [HR] = 0.47; 95% CI = 0.25–0.90; p = 0.023) (Fig. 1). In our study, the adjusted HR for an association of known variables for in-hospital proportion of death due to COVID-19 were 1.82 (95% CI = 1.01–3.27; p = 0.047) for hypertension and 3.25 (95% CI = 1.27–8.30; p = 0.014) for tuberculosis.

Table II.
Patient characteristics in tocilizumab group and nontocilizumab group after treatment
IndicatorsTotalTocilizumabNontocilizumabp Value
Outcome of disease     
 ICU admission (n = 195) 110 (56.41%) 40 (61.54%) 70 (53.85%) 0.307 
 Total days in ICU (n = 110) 11.00 (7.00–19.00) 10.00 (3.50–18.75) 12.00 (8.00–19.00) 0.271 
 Days from admission to discharge (n = 139) 29.00 (22.00–41.00) 40.00 (25.00–34.00) 26.50 (22.00–34.00) 0.001a 
 Nonsurvival (n = 195) 56 (28.72%) 14 (21.54%) 42 (32.31%) 0.117 
 Survival (n = 195) 139 (71.28%) 51 (78.46%) 88 (67.69%)  
Inflammatory cytokines and biomarkers     
 IL-6 (n = 195) in pg/ml 70.44 (31.62–450.85) 104.00 (38.18–515.40) 52.01 (29.97–440.78) 0.123 
 IL-8 (n = 180) in pg/l 19.10 (10.98–77.28) 16.45 (9.35–42.35) 21.40 (11.30–123.50) 0.201 
 IL-10 (n = 180) in pg/ml 6.30 (5.00–17.43) 5.00 (5.00–11.98) 7.35 (5.00–18.40) 0.013a 
 IL-1β (n = 180) in pg/ml 5.00 (5.00–8.45) 5.00 (5.00–8.48) 5.00 (5.00–8.45) 0.797 
 IL-2R (n = 179) in U/ml 790.00 (548.50–1300.50) 708.00 (507.25–1120.75) 839.00 (566.50–1324.00) 0.178 
 hs-CRP (n = 192) in mg/l 14.35 (1.65–83.38) 1.52 (0.40–21.63) 22.55 (4.68–111.50) <0.0001a 
 PCT (n = 171) in ng/ml 0.09 (0.05–0.82) 0.06 (0.04–0.60) 0.11 (0.05–0.90) 0.235 
Lymphocytes     
 Lymphocytes count (n = 194) (×109/l) 0.96 (0.54–1.47) 0.96 (0.51–1.27) 0.98 (0.54–1.54) 0.689 
 CD4+T cell count (n = 64) (/μl) 339.50 (237.25–495.75) 395.00 (276.51–644.00) 305.00 (174.50–429.00) 0.064 
 CD8+T cell count (n = 64) (/μl) 151.00 (53.75–316.50) 256.98 (149.50–356.26) 78.00 (39.25–256.75) 0.003a 
 CD3+T cell count (n = 64) (/μl) 422.03 (216.50–757.00) 608.29 (447.80–954.25) 300.00 (117.50–531.50) 0.001a 
 CD3CD16+ CD56+NK cell (n = 55) (/μl) 74.00 (22.50–139.50) 112.00 (66.00–158.00) 45.50 (16.75–127.50) 0.053 
 T cell+B cell+NK cell (n = 55) (/μl) 729.00 (451.00–1029.00) 834.00 (587.00–978.00) 665.50 (356.00–1052.50) 0.219 
 CD19+B cell (n = 55) (/μl) 117.00 (46.50–165.50) 113.00 (64.00–144.00) 119.50 (45.25–170.00) 0.689 
Blood routine     
 Neutrophils (n = 194) (×109/l) 5.23 (3.33–9.20) 5.93 (3.48–9.13) 4.67 (3.24–9.18) 0.521 
 Eosinophils (n = 194) (×109/l) 0.07 (0.01–0.18) 0.06 (0.00–0.14) 0.08 (0.02–0.18) 0.265 
 Basophils (n = 194) (×109/l) 0.02 (0.01–0.04) 0.02 (0.01–0.04) 0.02 (0.01–0.04) 0.131 
 Monocytes (n = 194) (×109/l) 0.48 (0.31–0.68) 0.46 (0.27–0.69) 0.48 (0.34–0.68) 0.847 
Organ damage indices     
 D-Dimer (n = 179) (μg/ml) 1.55 (0.63–3.33) 1.53 (0.73–2.95) 1.69 (0.62–3.88) 0.446 
 △D-Dimer (n = 179) (μg/ml) 0.00 (−1.33–0.86) −0.46 (−6.72 to 1.31) 0.00 (−078–0.71) 0.030a 
 APTT (n = 180) (s) 40.75 (35.08–47.05) 36.80 (34.70–42.10) 42.30 (37.10–48.35) 0.006a 
 PT (n = 181) (s) 14.20 (13.30–16.20) 13.90 (13.20–15.70) 14.35 (13.50–16.25) 0.050 
 Fbg (n = 179) (g/l) 4.16 (2.96–5.21) 2.57 (2.02–3.72) 4.71 (3.71–5.67) <0.0001a 
 ALT (n = 195) (U/l) 23.00 (15.00–36.00) 24.00 (17.00–37.00) 22.00 (15.00–36.00) 0.570 
 AST (n = 195) (U/l) 24.00 (19.00–40.00) 22.00 (19.00–40.00) 25.00 (19.00–40.75) 0.452 
 NT-proBNP (n = 163) (pg/ml) 346.50 (99.50–3660.00) 285.50 (107.75–3418.25) 446.00 (92.50–3683.00) 0.884 
 CK (n = 142) (U/l) 47.00 (30.00–108.25) 40.00 (30.00–59.75) 53.00 (29.25–179.75) 0.051 
 CK-MB (n = 135) (U/l) 1.10 (0.60–3.45) 1.00 (0.60–2.80) 1.10 (0.60–4.95) 0.925 
 hs-cTnI (n = 163) (μg/l) 10.40 (3.15–44.00) 5.60 (2.95–50.30) 11.95 (3.50–39.05) 0.301 
IndicatorsTotalTocilizumabNontocilizumabp Value
Outcome of disease     
 ICU admission (n = 195) 110 (56.41%) 40 (61.54%) 70 (53.85%) 0.307 
 Total days in ICU (n = 110) 11.00 (7.00–19.00) 10.00 (3.50–18.75) 12.00 (8.00–19.00) 0.271 
 Days from admission to discharge (n = 139) 29.00 (22.00–41.00) 40.00 (25.00–34.00) 26.50 (22.00–34.00) 0.001a 
 Nonsurvival (n = 195) 56 (28.72%) 14 (21.54%) 42 (32.31%) 0.117 
 Survival (n = 195) 139 (71.28%) 51 (78.46%) 88 (67.69%)  
Inflammatory cytokines and biomarkers     
 IL-6 (n = 195) in pg/ml 70.44 (31.62–450.85) 104.00 (38.18–515.40) 52.01 (29.97–440.78) 0.123 
 IL-8 (n = 180) in pg/l 19.10 (10.98–77.28) 16.45 (9.35–42.35) 21.40 (11.30–123.50) 0.201 
 IL-10 (n = 180) in pg/ml 6.30 (5.00–17.43) 5.00 (5.00–11.98) 7.35 (5.00–18.40) 0.013a 
 IL-1β (n = 180) in pg/ml 5.00 (5.00–8.45) 5.00 (5.00–8.48) 5.00 (5.00–8.45) 0.797 
 IL-2R (n = 179) in U/ml 790.00 (548.50–1300.50) 708.00 (507.25–1120.75) 839.00 (566.50–1324.00) 0.178 
 hs-CRP (n = 192) in mg/l 14.35 (1.65–83.38) 1.52 (0.40–21.63) 22.55 (4.68–111.50) <0.0001a 
 PCT (n = 171) in ng/ml 0.09 (0.05–0.82) 0.06 (0.04–0.60) 0.11 (0.05–0.90) 0.235 
Lymphocytes     
 Lymphocytes count (n = 194) (×109/l) 0.96 (0.54–1.47) 0.96 (0.51–1.27) 0.98 (0.54–1.54) 0.689 
 CD4+T cell count (n = 64) (/μl) 339.50 (237.25–495.75) 395.00 (276.51–644.00) 305.00 (174.50–429.00) 0.064 
 CD8+T cell count (n = 64) (/μl) 151.00 (53.75–316.50) 256.98 (149.50–356.26) 78.00 (39.25–256.75) 0.003a 
 CD3+T cell count (n = 64) (/μl) 422.03 (216.50–757.00) 608.29 (447.80–954.25) 300.00 (117.50–531.50) 0.001a 
 CD3CD16+ CD56+NK cell (n = 55) (/μl) 74.00 (22.50–139.50) 112.00 (66.00–158.00) 45.50 (16.75–127.50) 0.053 
 T cell+B cell+NK cell (n = 55) (/μl) 729.00 (451.00–1029.00) 834.00 (587.00–978.00) 665.50 (356.00–1052.50) 0.219 
 CD19+B cell (n = 55) (/μl) 117.00 (46.50–165.50) 113.00 (64.00–144.00) 119.50 (45.25–170.00) 0.689 
Blood routine     
 Neutrophils (n = 194) (×109/l) 5.23 (3.33–9.20) 5.93 (3.48–9.13) 4.67 (3.24–9.18) 0.521 
 Eosinophils (n = 194) (×109/l) 0.07 (0.01–0.18) 0.06 (0.00–0.14) 0.08 (0.02–0.18) 0.265 
 Basophils (n = 194) (×109/l) 0.02 (0.01–0.04) 0.02 (0.01–0.04) 0.02 (0.01–0.04) 0.131 
 Monocytes (n = 194) (×109/l) 0.48 (0.31–0.68) 0.46 (0.27–0.69) 0.48 (0.34–0.68) 0.847 
Organ damage indices     
 D-Dimer (n = 179) (μg/ml) 1.55 (0.63–3.33) 1.53 (0.73–2.95) 1.69 (0.62–3.88) 0.446 
 △D-Dimer (n = 179) (μg/ml) 0.00 (−1.33–0.86) −0.46 (−6.72 to 1.31) 0.00 (−078–0.71) 0.030a 
 APTT (n = 180) (s) 40.75 (35.08–47.05) 36.80 (34.70–42.10) 42.30 (37.10–48.35) 0.006a 
 PT (n = 181) (s) 14.20 (13.30–16.20) 13.90 (13.20–15.70) 14.35 (13.50–16.25) 0.050 
 Fbg (n = 179) (g/l) 4.16 (2.96–5.21) 2.57 (2.02–3.72) 4.71 (3.71–5.67) <0.0001a 
 ALT (n = 195) (U/l) 23.00 (15.00–36.00) 24.00 (17.00–37.00) 22.00 (15.00–36.00) 0.570 
 AST (n = 195) (U/l) 24.00 (19.00–40.00) 22.00 (19.00–40.00) 25.00 (19.00–40.75) 0.452 
 NT-proBNP (n = 163) (pg/ml) 346.50 (99.50–3660.00) 285.50 (107.75–3418.25) 446.00 (92.50–3683.00) 0.884 
 CK (n = 142) (U/l) 47.00 (30.00–108.25) 40.00 (30.00–59.75) 53.00 (29.25–179.75) 0.051 
 CK-MB (n = 135) (U/l) 1.10 (0.60–3.45) 1.00 (0.60–2.80) 1.10 (0.60–4.95) 0.925 
 hs-cTnI (n = 163) (μg/l) 10.40 (3.15–44.00) 5.60 (2.95–50.30) 11.95 (3.50–39.05) 0.301 

Sixty-five patients using tocilizumab and 130 patients not taking tocilizumab were compared. Laboratory and clinical outcomes data after treatment of all patients with laboratory-confirmed SARS-CoV-2 were obtained with data collection forms from electronic medical records of Tongji Hospital, Wuhan Pulmonary Hospital and Renmin Hospital of Wuhan University (the east campus). Continuous variables were described as median and IQR or mean and SD, and differences were assessed using analysis of Student t test or Mann–Whitney U test. Categorical variables were expressed as number (percentage), and differences between groups were assessed using Pearson χ2 test, Fisher exact test, or the continuity correction χ2 tests.

a

Data shown are significant at p < 0.05.

Abbreviations used in this table: CD, cluster of differentiation; CK, creatine kinase; CK-MB, creatine kinase–myocardial band; Fbg, fibrinogen; hs-CRP, hypersensitive CRP; NT-proBNP, N-terminal brain natriuretic propeptide; PCT, procalcitonin; PT, plasma prothrombin time.

FIGURE 1.

Survival of COVID-19 patients during hospitalization. Kaplan–Meier curves for cumulative p value of COVID-19 mortality during follow up duration in tocilizumab or non-tocilizumab cohort among 195 patients within Cox proportional hazards model.

FIGURE 1.

Survival of COVID-19 patients during hospitalization. Kaplan–Meier curves for cumulative p value of COVID-19 mortality during follow up duration in tocilizumab or non-tocilizumab cohort among 195 patients within Cox proportional hazards model.

Close modal

Total days in the ICU in the tocilizumab group are shorter than those in the nontocilizumab group (10.00 versus 12.00 d; p = 0.27), and the duration from admission to hospital discharge was significantly longer (40.00 versus 26.50 d; p = 0.001) (Table II).

SARS-CoV-2 infection can arouse both pulmonary and multisystem inflammation, leading to critical complications. In this study, we observed that the incidence of ARDS (36.92 versus 70.77%; p < 0.0001) was lower in the tocilizumab group than in the nontocilizumab group (Table II). In the multivariable logistic regression models, use of tocilizumab was associated with lower risk of ARDS (OR = 0.23; 95% CI = 0.11–0.45; p < 0.0001) (Table III). These data suggest that IL-6 may be a potential actionable target cytokine to treat COVID-19–related ARDS and that tocilizumab is effective in reducing the incidence of ARDS.

Table III.
HR for outcomes in tocilizumab group versus nontocilizumab group
Univariable Logistic RegressionMultivariable Logistic Regression
ComplicationsOR (95% CI)p ValueOR (95% CI)p Value
ARDS 0.24 (0.13–0.45) <0.0001a 0.23 (0.11–0.45) <0.0001a 
DIC 0.28 (0.03–2.28) 0.231 0.26 (0.03–2.37) 0.232 
Acute renal injury 0.78 (0.39–1.57) 0.49 0.69 (0.31–1.51) 0.35 
Acute liver injury 1.63 (0.78–3.32) 0.203 1.75 (0.78–3.92) 0.175 
Acute cardiac injury 1.14 (0.62–2.09) 0.679 0.98 (0.49–1.98) 0.960 
Univariable Logistic RegressionMultivariable Logistic Regression
ComplicationsOR (95% CI)p ValueOR (95% CI)p Value
ARDS 0.24 (0.13–0.45) <0.0001a 0.23 (0.11–0.45) <0.0001a 
DIC 0.28 (0.03–2.28) 0.231 0.26 (0.03–2.37) 0.232 
Acute renal injury 0.78 (0.39–1.57) 0.49 0.69 (0.31–1.51) 0.35 
Acute liver injury 1.63 (0.78–3.32) 0.203 1.75 (0.78–3.92) 0.175 
Acute cardiac injury 1.14 (0.62–2.09) 0.679 0.98 (0.49–1.98) 0.960 

Sixty-five patients using tocilizumab and 130 patients not taking tocilizumab were compared. Complication data after treatment of all patients with laboratory-confirmed SARS-CoV-2 were obtained with data collection forms from electronic medical records of Tongji Hospital, Wuhan Pulmonary Hospital, and Renmin Hospital of Wuhan University (the east campus). The multivariable logistic regression model was adjusted for age, sex, and comorbidities. ORs and 95% CIs were calculated by univariable logistic regression models.

a

Data shown are significant at p < 0.05.

Considering that CRS refers to an uncontrolled and overwhelming release of proinflammatory mediators by an overly activated immune system, we analyzed the difference between the tocilizumab group and the nontocilizumab group based on the levels of inflammatory cytokines and immune cells subsets after treatment (Table II). Analysis revealed that the infection-related biomarkers, including IL-10 (5.00 versus 7.35 pg/ml; p = 0.013) and C-reactive protein (CRP; 1.52 versus 22.55 mg/l; p < 0.0001) were remarkably decreased in the tocilizumab group compared with the nontocilizumab group (Table II). Consistent with recently published studies, results revealed that CRS-related cytokines, especially CRP, were rapidly reduced in patients with severe COVID-19 after receiving tocilizumab (20, 21). In contrast, the count of CD3+ T cells (608.29 versus 300.00/μl; p = 0.001) was significantly increased in tocilizumab group than that in the nontocilizumab group (Table II). For further analysis of T cell subsets, we found that the count of CD4+ T cells (395.00 versus 305.00/μl; p = 0.064) and CD8+ T cells (256.98 versus 78.00/μl; p = 0.003) was increased as well as the total number of T cells (Table II). These findings indicate that COVID-19 patients using tocilizumab have more improved inflammation and immune cell function recovery than those who did not. The dynamic change of four indices over follow-up are shown in the Fig. 2. After the patient was admitted to the hospital, the level of CRP increased, whereas CD3+ T cell and CD8+ T cell counts declined in both groups as the disease progressed. After tocilizumab treatment, CRP decreased from 60.85 to 1.52 mg/l in tocilizumab group, and the number of immune cells was higher than the baseline, especially in the tocilizumab group. The difference did not reach statistical significance, but it presented a statistical trend, which may be attributed to the limited sample size and short follow-up time. Noticeably, we observed that CRP and T cell numbers appeared as a significant change in tocilizumab group at week 7 compared with the control group. It is warranted to evaluate the effect of tocilizumab in larger scale prospective cohort studies and randomized controlled trials.

FIGURE 2.

Temporal changes in laboratory parameters in COVID-19 patients after admission. The temporal changes of hypersensitive CRP (hs-CRP) (A), CD3+ T cell (B), CD8+ T cell (C), and lymphocyte (D) in tocilizumab group and nontocilizumab group. These parameters were measured at 1, 2, 3, 4, 5, 6, and 7 wk after admission. The green dotted line represents the lower limit of normal, and the red dotted line represents the upper limit of normal. The levels of these parameters at each point were expressed as median and IQR. **p < 0.01, ***p < 0.001 were calculated by Mann–Whitney U test for last examination.

FIGURE 2.

Temporal changes in laboratory parameters in COVID-19 patients after admission. The temporal changes of hypersensitive CRP (hs-CRP) (A), CD3+ T cell (B), CD8+ T cell (C), and lymphocyte (D) in tocilizumab group and nontocilizumab group. These parameters were measured at 1, 2, 3, 4, 5, 6, and 7 wk after admission. The green dotted line represents the lower limit of normal, and the red dotted line represents the upper limit of normal. The levels of these parameters at each point were expressed as median and IQR. **p < 0.01, ***p < 0.001 were calculated by Mann–Whitney U test for last examination.

Close modal

We also evaluated the dynamic changes of organ damage indices at final examination (Table II). Biomarkers of multiple organ dysfunction in the tocilizumab group were within the normal range. After the treatment with tocilizumab, the activated coagulation system by IL-6 returned to normal. Activated partial thromboplastin time (APTT) was shorter than in the nontocilizumab group (36.80 versus 42.30 s; p = 0.006), and the decline of fibrinogen (2.57 versus 4.71 g/l; p < 0.0001) was more pronounced in tocilizumab group (Table II).

The percentages of patients with adverse reactions were similar in the two groups; there were significant differences (Supplemental Table IV). The most common adverse event in the tocilizumab group was increasing liver enzymes (AST, 8/65 [12.31%]; ALT, 9/65 [13.85%]) as previously reported (22).

In this multicenter retrospective study involving a modest sample of consecutive patients who had been hospitalized with COVID-19, the risk of in-hospital proportion of death was significantly lower among patients who received tocilizumab than among those who did not. It revealed that tocilizumab could prolong survival among patients with COVID-19, specifically among hospitalized COVID-19 patients. Clinical data showed that tocilizumab treatment weakens the inflammatory response while sustaining immune responses in posttreatment COVID-19 patients, suggesting that tocilizumab could be an efficient therapeutic for the treatment of COVID-19. Although it is plausible that unmeasured confounding factors may have contributed to the observed protective association, these data suggested that in-hospital use of tocilizumab was not associated with increased proportion of death in COVID-19. These findings provide clinical evidence in support of recently published guidance statements by National Health Commission of the People’s Republic of China to continue tocilizumab in patients with severe or critical COVID-19 (14, 23). Given the observational design, whether tocilizumab could reduce overall mortality in patients with COVID-19 needs further validation in geographically diverse, large-scale prospective cohort studies.

CRS was found to be the major cause of morbidity in patients infected with severe SARS-CoV and MERS-CoV (8). Elevated serum concentrations of the IL-6 are hallmarks of severe MERS-CoV infections (24). Elevated serum CRP, a protein whose expression is driven by IL-6, is also a biomarker of severe β-coronavirus infection. Recent studies have also reported that hypercytokinemia is associated with a higher proportion of death in patients with severe COVID-19. IL-6 is an excellent biomarker of severity and a prognostic indicator for most of diseases presenting with a cytokine storm and is expressed longer than TNF-α and IL-1 (25). Although IL-6 is regulated strictly through transcription and posttranscription mechanisms, dysregulated, persistent production of IL-6 plays a pathological role in tissue hypoxia, hypotension, DIC, and multiple organ dysfunction (2629). Mechanistically, IL-6 is essential for the generation of Th17 cells in the dendritic cell–T cell interaction (30). The excessive IL-6 may explain the overly activated Th17 cells observed in COVID-19 patients (31). A high level of IL-6 is associated with poor outcome in the setting of COVID-19 pneumonia (32). Accumulating evidence evoked the use of tocilizumab in COVID-19 infection. Tocilizumab, an IL-6R monoclonal blocking agent, was approved for the treatment of rheumatoid arthritis in Japan (2008), Europe (2009), and the United States (2010) (4, 3335). Mechanistically, tocilizumab can bind both soluble and membrane-bound IL-6R to inhibit IL-6–mediated cis- and trans-signaling (10). A new study found that the inhibition of IL-6 signaling by tocilizumab treatment decreased PAI-1 production and improved the vascular endothelial functions in patients with severe COVID-19. IL-6 trans-signaling forms a positive feedback loop and plays pivotal roles in endothelial cell injury and coagulopathy, thereby mediating systemic inflammation and deteriorating systemic circulation (36). A phase II trial is ongoing in Italy (37), and a phase III trial was approved by the Food and Drug Administration (38) to assess the effect of tocilizumab for severe COVID-19 pneumonia. More than 1 million patients worldwide have been treated with tocilizumab, and clinical trials have proved that tocilizumab is safe for both pediatric and adult patients (22). In the mainly rheumatologic indications, the tolerance of tocilizumab is generally good, the main adverse events are a transient decrease in leukocytes, an increase in liver enzymes, and a slight increase of bacterial infection (22).

In COVID-19 patients with elevated inflammatory cytokines, postmortem pathology has revealed tissue necrosis and interstitial macrophage and monocyte infiltrations in the lung, heart, and gastrointestinal mucosa (22). Moreover, severe lymphopenia with hyperactivated proinflammatory T cells and decreased regulatory T cells are commonly seen in critically ill patients, suggesting dysregulated immune responses. In our study, 65 patients had a history of routine treatment for ∼15 days before tocilizumab treatment. A lower percentage of lymphocytes was found in patients, and it returned to normal when tocilizumab was given. At the same time, patients deteriorated with increased inflammatory response (inflammatory cytokines were elevated [e.g., IL-6, CRP]) and decreased immune function (immune cells decreased [e.g., CD4+ T cell, CD8+ T cell]) before tocilizumab. After the treatment, in addition to partially rescuing the immune dysregulation, the inflammatory response was weakened to some degrees in the patients. During the treatment, no severe adverse drug reactions were reported. Clinical symptoms of all patients improved with good prognosis after the treatment. Overall, these findings suggested that tocilizumab group seemed to have an improved survival outcome that might be explained by the blocking of IL-6–associated dysregulated immune responses and inflammatory storm response.

Our study has several limitations. First, this retrospective study population is derived from three hospitals in Hubei Province, but the effects of tocilizumab may vary among people of different races or geographic locations, which seems reasonable. Second, the potential mechanisms of dysregulated inflammatory cytokines and immune responses were not fully explored and need to be further investigated. Third, the sample size of the study was modest and included 65 patients who received tocilizumab. Currently, the sample size enrolled in case-control study was the largest. Because unmeasured biases are particular problems inherent in retrospective studies, we adjusted for likely confounders, including age, sex, and comorbidities in our analysis. Despite this extensive adjustment, it is still possible that some amount of unmeasured confounding remains. Therefore, large-scale prospective cohort studies and randomized controlled trials are needed to better understand the efficacy of tocilizumab in COVID-19. Recently a randomized, double-blind, placebo-controlled trial indicated that tocilizumab had no significant effect on the risk of intubation or death (39). However, this study states that because of the width of the CIs for efficacy comparisons, it cannot exclude the possibility that tocilizumab is associated with either some benefit or harm in some patients. Reported but still unpublished results of a small number of other randomized trials show that time to hospital discharge was shorter in patients treated with Actemra/RoActemra (tocilizumab) than in those treated with placebo (40). At the same time, our results suggest that tocilizumab has a week effect on survival among patients with COVID-19, specifically hospitalized COVID-19 patients. It is worth noting the limitations of our research; larger scale prospective cohort studies and extended follow-up time are warranted.

In summary, tocilizumab effectively improves clinical symptoms and represses the deterioration of patients with severe COVID-19. In our study, inpatient treatment with tocilizumab was associated with a lower risk of in-hospital proportion of death compared with tocilizumab nonusers. Although study interpretation needs to consider the potential for residual confounders, it is unlikely that inpatient tocilizumab would be associated with an increased risk of mortality. Noticeably, these results are consistent with those reported by other investigators (Refs. 41 and 42, N. Wadud, N. Ahmed, M. Mannu Shergil, M. Khan, M.G. Krishna, A. Gilani, S. El Zarif, J. Galaydick, K. Linga, S. Koor, et al., manuscript posted on medRxiv, DOI: 10.1101/2020.05.13.20100081; and C.A. Rimland, C.E. Morgan, G.J. Bell, M.K. Kim, T. Hedrick, A. Marx, B. Bramson, H. Swygard, S. Napravnik, J.L. Schmitz, et al., manuscript posted on medRxiv, DOI: 10.1101/2020.05.13.20100404), suggesting that these findings can be extrapolated to other populations or countries. Therefore, tocilizumab appears to be an effective treatment in patients with severe COVID-19, which provides a new therapeutic strategy for this fatal infectious disease.

We acknowledge all patients and families involved in the study and all health care workers who are fighting the campaign against COVID-19.

This work was supported by the SARS-CoV-2 Pneumonia Emergency Technology Public Relations Project of Tongji Medical College, Huazhong University of Science and Technology (Grant 2020kfyXGYJ043) and the National Key Research and Development Plan for the Emergency Management of Novel Coronavirus Pneumonia, China (Grant 2020YFC0845100).

The online version of this article contains supplemental material.

Abbreviations used in this article:

ALT

alanine transaminase

APTT

activated partial thromboplastin time

ARDS

acute respiratory distress syndrome

AST

aspartate aminotransferase

CI

confidence interval

COVID-19

coronavirus disease 2019

CRP

C-reactive protein

CRS

cytokine release syndrome

CT

computed tomography

DIC

disseminated intravascular coagulation

HR

hazard ratio

hs-cTnI

high-sensitivity cardiac troponin I

ICU

intensive care unit

IQR

interquartile range

OR

odds ratio

SARS-CoV-2

severe acute respiratory syndrome coronavirus 2.

1
Coronaviridae Study Group of the International Committee on Taxonomy of Viruses
.
2020
.
The species severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2.
Nat. Microbiol.
5
:
536
544
.
2
WHO. Coronavirus disease 2019 (COVID-19) situation report. Available at: https://www.who.int/emergencies/diseases/novel-coronavirus-2019. Accessed August 18, 2020.
3
Yang
,
X.
,
Y.
Yu
,
J.
Xu
,
H.
Shu
,
J.
Xia
,
H.
Liu
,
Y.
Wu
,
L.
Zhang
,
Z.
Yu
,
M.
Fang
, et al
.
2020
.
Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. [Published erratum appears in 2020 Lancet Respir. Med. 8: e26.]
Lancet Respir. Med.
8
:
475
481
.
4
Zhang
,
C.
,
Z.
Wu
,
J. W.
Li
,
H.
Zhao
,
G. Q.
Wang
.
2020
.
Cytokine release syndrome in severe COVID-19: interleukin-6 receptor antagonist tocilizumab may be the key to reduce mortality.
Int. J. Antimicrob. Agents
55
: 105954.
5
Moore
,
J. B.
,
C. H.
June
.
2020
.
Cytokine release syndrome in severe COVID-19.
Science
368
:
473
474
.
6
Du
,
M.
,
G.
Cai
,
F.
Chen
,
D. C.
Christiani
,
Z.
Zhang
,
M.
Wang
.
2020
.
Multiomics evaluation of gastrointestinal and other clinical characteristics of COVID-19.
Gastroenterology
158
:
2298
2301.e7
.
7
Borthwick
,
E. M.
,
C. J.
Hill
,
K. S.
Rabindranath
,
A. P.
Maxwell
,
D. F.
McAuley
,
B.
Blackwood
.
2017
.
High-volume haemofiltration for sepsis in adults.
Cochrane Database Syst. Rev.
1
: CD008075.
8
Channappanavar
,
R.
,
S.
Perlman
.
2017
.
Pathogenic human coronavirus infections: causes and consequences of cytokine storm and immunopathology.
Semin. Immunopathol.
39
:
529
539
.
9
Johnson
,
D. E.
,
R. A.
O’Keefe
,
J. R.
Grandis
.
2018
.
Targeting the IL-6/JAK/STAT3 signalling axis in cancer.
Nat. Rev. Clin. Oncol.
15
:
234
248
.
10
Le
,
R. Q.
,
L.
Li
,
W.
Yuan
,
S. S.
Shord
,
L.
Nie
,
B. A.
Habtemariam
,
D.
Przepiorka
,
A. T.
Farrell
,
R.
Pazdur
.
2018
.
FDA approval summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or life-threatening cytokine release syndrome.
Oncologist
23
:
943
947
.
11
Choy
,
E. H.
,
F.
De Benedetti
,
T.
Takeuchi
,
M.
Hashizume
,
M. R.
John
,
T.
Kishimoto
.
2020
.
Translating IL-6 biology into effective treatments.
Nat. Rev. Rheumatol.
16
:
335
345
.
12
Neelapu
,
S. S.
,
S.
Tummala
,
P.
Kebriaei
,
W.
Wierda
,
C.
Gutierrez
,
F. L.
Locke
,
K. V.
Komanduri
,
Y.
Lin
,
N.
Jain
,
N.
Daver
, et al
.
2018
.
Chimeric antigen receptor T-cell therapy - assessment and management of toxicities.
Nat. Rev. Clin. Oncol.
15
:
47
62
.
13
Veselka
,
J.
,
L.
Faber
,
M.
Liebregts
,
R.
Cooper
,
J.
Januska
,
M.
Kashtanov
,
M.
Dabrowski
,
P. R.
Hansen
,
H.
Seggewiss
,
E.
Hansvenclova
, et al
.
2019
.
Short- and long-term outcomes of alcohol septal ablation for hypertrophic obstructive cardiomyopathy in patients with mild left ventricular hypertrophy: a propensity score matching analysis.
Eur. Heart J.
40
:
1681
1687
.
14
Zhou
,
F.
,
T.
Yu
,
R.
Du
,
G.
Fan
,
Y.
Liu
,
Z.
Liu
,
J.
Xiang
,
Y.
Wang
,
B.
Song
,
X.
Gu
, et al
.
2020
.
Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. [Published erratum appears in 2020 Lancet 395: 1038.]
Lancet
395
:
1054
1062
.
15
Pan
,
A.
,
L.
Liu
,
C.
Wang
,
H.
Guo
,
X.
Hao
,
Q.
Wang
,
J.
Huang
,
N.
He
,
H.
Yu
,
X.
Lin
, et al
.
2020
.
Association of public health interventions with the epidemiology of the COVID-19 outbreak in Wuhan, China.
JAMA
323
:
1915
1923
.
16
Auriemma
,
C. L.
,
H.
Zhuo
,
K.
Delucchi
,
T.
Deiss
,
T.
Liu
,
A.
Jauregui
,
S.
Ke
,
K.
Vessel
,
M.
Lippi
,
E.
Seeley
, et al
.
2020
.
Acute respiratory distress syndrome-attributable mortality in critically ill patients with sepsis.
Intensive Care Med.
46
:
1222
1231
.
17
Levi
,
M.
,
M.
Scully
.
2018
.
How I treat disseminated intravascular coagulation.
Blood
131
:
845
854
.
18
Kellum
,
J. A.
,
N.
Lameire
;
KDIGO AKI Guideline Work Group
.
2013
.
Diagnosis, evaluation, and management of acute kidney injury: a KDIGO summary (Part 1).
Crit. Care
17
:
204
.
19
Huang
,
C.
,
Y.
Wang
,
X.
Li
,
L.
Ren
,
J.
Zhao
,
Y.
Hu
,
L.
Zhang
,
G.
Fan
,
J.
Xu
,
X.
Gu
, et al
.
2020
.
Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. [Published erratum appears in 2020 Lancet 395: 496.]
Lancet
395
:
497
506
.
20
Wang
,
D.
,
B.
Hu
,
C.
Hu
,
F.
Zhu
,
X.
Liu
,
J.
Zhang
,
B.
Wang
,
H.
Xiang
,
Z.
Cheng
,
Y.
Xiong
, et al
.
2020
.
Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China.
JAMA
323
:
1061
1069
.
21
Chen
,
G.
,
D.
Wu
,
W.
Guo
,
Y.
Cao
,
D.
Huang
,
H.
Wang
,
T.
Wang
,
X.
Zhang
,
H.
Chen
,
H.
Yu
, et al
.
2020
.
Clinical and immunological features of severe and moderate coronavirus disease 2019.
J. Clin. Invest.
130
:
2620
2629
.
22
Burmester
,
G. R.
,
A.
Rubbert-Roth
,
A.
Cantagrel
,
S.
Hall
,
P.
Leszczynski
,
D.
Feldman
,
M. J.
Rangaraj
,
G.
Roane
,
C.
Ludivico
,
M.
Bao
, et al
.
2016
.
Efficacy and safety of subcutaneous tocilizumab versus intravenous tocilizumab in combination with traditional DMARDs in patients with RA at week 97 (SUMMACTA).
Ann. Rheum. Dis.
75
:
68
74
.
23
Pan
,
A.
,
L.
Liu
,
C.
Wang
,
H.
Guo
,
X.
Hao
,
Q.
Wang
,
J.
Huang
,
N.
He
,
H.
Yu
,
X.
Lin
, et al
.
2020
.
Association of public health interventions with the epidemiology of the COVID-19 outbreak in Wuhan, China.
JAMA
323
:
1915
1923
.
24
Fehr
,
A. R.
,
R.
Channappanavar
,
S.
Perlman
.
2017
.
Middle East respiratory syndrome: emergence of a pathogenic human coronavirus.
Annu. Rev. Med.
68
:
387
399
.
25
Damas
,
P.
,
D.
Ledoux
,
M.
Nys
,
Y.
Vrindts
,
D.
De Groote
,
P.
Franchimont
,
M.
Lamy
.
1992
.
Cytokine serum level during severe sepsis in human IL-6 as a marker of severity.
Ann. Surg.
215
:
356
362
.
26
Tanaka
,
T.
,
T.
Kishimoto
.
2014
.
The biology and medical implications of interleukin-6.
Cancer Immunol. Res.
2
:
288
294
.
27
Stouthard
,
J. M.
,
M.
Levi
,
C. E.
Hack
,
C. H.
Veenhof
,
H. A.
Romijn
,
H. P.
Sauerwein
,
T.
van der Poll
.
1996
.
Interleukin-6 stimulates coagulation, not fibrinolysis, in humans.
Thromb. Haemost.
76
:
738
742
.
28
Romano
,
M.
,
M.
Sironi
,
C.
Toniatti
,
N.
Polentarutti
,
P.
Fruscella
,
P.
Ghezzi
,
R.
Faggioni
,
W.
Luini
,
V.
van Hinsbergh
,
S.
Sozzani
, et al
.
1997
.
Role of IL-6 and its soluble receptor in induction of chemokines and leukocyte recruitment.
Immunity
6
:
315
325
.
29
Lo
,
C. W.
,
M. W.
Chen
,
M.
Hsiao
,
S.
Wang
,
C. A.
Chen
,
S. M.
Hsiao
,
J. S.
Chang
,
T. C.
Lai
,
S.
Rose-John
,
M. L.
Kuo
,
L. H.
Wei
.
2011
.
IL-6 trans-signaling in formation and progression of malignant ascites in ovarian cancer.
Cancer Res.
71
:
424
434
.
30
Tanaka
,
T.
,
M.
Narazaki
,
T.
Kishimoto
.
2016
.
Immunotherapeutic implications of IL-6 blockade for cytokine storm.
Immunotherapy
8
:
959
970
.
31
Xu
,
Z.
,
L.
Shi
,
Y.
Wang
,
J.
Zhang
,
L.
Huang
,
C.
Zhang
,
S.
Liu
,
P.
Zhao
,
H.
Liu
,
L.
Zhu
, et al
.
2020
.
Pathological findings of COVID-19 associated with acute respiratory distress syndrome.
Lancet Respir. Med.
8
:
420
422
.
32
Antinori
,
S.
,
C.
Bonazzetti
,
G.
Gubertini
,
A.
Capetti
,
C.
Pagani
,
V.
Morena
,
S.
Rimoldi
,
L.
Galimberti
,
P.
Sarzi-Puttini
,
A. L.
Ridolfo
.
2020
.
Tocilizumab for cytokine storm syndrome in COVID-19 pneumonia: an increased risk for candidemia?
Autoimmun. Rev.
19
: 102564.
33
Mehta
,
P.
,
D. F.
McAuley
,
M.
Brown
,
E.
Sanchez
,
R. S.
Tattersall
,
J. J.
Manson
;
HLH Across Speciality Collaboration, UK
.
2020
.
COVID-19: consider cytokine storm syndromes and immunosuppression.
Lancet
395
:
1033
1034
.
34
Ogata
,
A.
,
Y.
Kato
,
S.
Higa
,
K.
Yoshizaki
.
2019
.
IL-6 inhibitor for the treatment of rheumatoid arthritis: a comprehensive review.
Mod. Rheumatol.
29
:
258
267
.
35
Wu
,
C.
,
X.
Chen
,
Y.
Cai
,
J.
Xia
,
X.
Zhou
,
S.
Xu
,
H.
Huang
,
L.
Zhang
,
X.
Zhou
,
C.
Du
, et al
.
2020
.
Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China.
JAMA Intern. Med.
180
:
934
943
.
36
Kang
,
S.
,
T.
Tanaka
,
H.
Inoue
,
C.
Ono
,
S.
Hashimoto
,
Y.
Kioi
,
H.
Matsumoto
,
H.
Matsuura
,
T.
Matsubara
,
K.
Shimizu
, et al
.
2020
.
IL-6 trans-signaling induces plasminogen activator inhibitor-1 from vascular endothelial cells in cytokine release syndrome.
Proc. Natl. Acad. Sci. USA
117
:
22351
22356
.
37
US National Library of Medicine. ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04317092. Accessed November 4, 2020.
38
US National Library of Medicine. ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04320615. Accessed November 4, 2020.
39
Stone, J. H., M. J. Frigault, N. J. Serling-Boyd, A. D. Fernandes, L. Harvey, A. S. Foulkes, N. K. Horick, B. C. Healy, R. Shah, A. M. Bensaci, et al. 2020. Efficacy of tocilizumab in patients hospitalized with covid-19. N. Engl. J. Med. DOI: 10.1056/NEJMoa2028836.
40
Roche
.
2020
. Roche provides an update on the phase III COVACTA trial of Actemra/RoActemra in hospitalised patients with severe COVID-19 associated pneumonia. Available at: https://www.roche.com/investors/updates/inv-update-2020-07-29.htm.
41
Xu
,
X.
,
M.
Han
,
T.
Li
,
W.
Sun
,
D.
Wang
,
B.
Fu
,
Y.
Zhou
,
X.
Zheng
,
Y.
Yang
,
X.
Li
, et al
.
2020
.
Effective treatment of severe COVID-19 patients with tocilizumab.
Proc. Natl. Acad. Sci. USA
117
:
10970
10975
.
42
Capra
,
R.
,
N.
De Rossi
,
F.
Mattioli
,
G.
Romanelli
,
C.
Scarpazza
,
M. P.
Sormani
,
S.
Cossi
.
2020
.
Impact of low dose tocilizumab on mortality rate in patients with COVID-19 related pneumonia.
Eur. J. Intern. Med.
76
:
31
35
.

The authors have no financial conflicts of interest.

This article is distributed under The American Association of Immunologists, Inc., Reuse Terms and Conditions for Author Choice articles.

Supplementary data