Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Can we predict the severe course of COVID-19 - a systematic review and meta-analysis of indicators of clinical outcome?

  • Stephan Katzenschlager ,

    Contributed equally to this work with: Stephan Katzenschlager, Alexandra J. Zimmer

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Project administration, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany

  • Alexandra J. Zimmer ,

    Contributed equally to this work with: Stephan Katzenschlager, Alexandra J. Zimmer

    Roles Formal analysis, Methodology, Software, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Departments of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada

  • Claudius Gottschalk,

    Roles Conceptualization, Data curation, Methodology, Writing – review & editing

    Affiliation Division of Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany

  • Jürgen Grafeneder,

    Roles Conceptualization, Data curation, Methodology, Writing – review & editing

    Affiliation Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria

  • Stephani Schmitz,

    Roles Data curation, Formal analysis

    Affiliation Division of Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany

  • Sara Kraker,

    Roles Data curation, Formal analysis

    Affiliation Division of Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany

  • Marlene Ganslmeier,

    Roles Data curation, Formal analysis

    Affiliation Division of Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany

  • Amelie Muth,

    Roles Data curation, Formal analysis

    Affiliation Division of Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany

  • Alexander Seitel,

    Roles Conceptualization, Data curation, Methodology, Writing – review & editing

    Affiliation Division of Computer Assisted Medical Interventions, German Cancer Research Center (DKFZ), Heidelberg, Germany

  • Lena Maier-Hein,

    Roles Data curation, Methodology, Software, Writing – review & editing

    Affiliation Division of Computer Assisted Medical Interventions, German Cancer Research Center (DKFZ), Heidelberg, Germany

  • Andrea Benedetti,

    Roles Conceptualization, Methodology, Writing – review & editing

    Affiliation Departments of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada

  • Jan Larmann,

    Roles Writing – review & editing

    Affiliation Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany

  • Markus A. Weigand,

    Roles Writing – review & editing

    Affiliation Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany

  • Sean McGrath ,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Software, Writing – original draft, Writing – review & editing

    ‡ These authors also contributed equally to this work.

    Affiliation Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America

  • Claudia M. Denkinger

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing

    Claudia.Denkinger@uni-heidelberg.de

    ‡ These authors also contributed equally to this work.

    Affiliations Division of Tropical Medicine, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany, German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany

Abstract

Background

COVID-19 has been reported in over 40million people globally with variable clinical outcomes. In this systematic review and meta-analysis, we assessed demographic, laboratory and clinical indicators as predictors for severe courses of COVID-19.

Methods

This systematic review was registered at PROSPERO under CRD42020177154. We systematically searched multiple databases (PubMed, Web of Science Core Collection, MedRvix and bioRvix) for publications from December 2019 to May 31st 2020. Random-effects meta-analyses were used to calculate pooled odds ratios and differences of medians between (1) patients admitted to ICU versus non-ICU patients and (2) patients who died versus those who survived. We adapted an existing Cochrane risk-of-bias assessment tool for outcome studies.

Results

Of 6,702 unique citations, we included 88 articles with 69,762 patients. There was concern for bias across all articles included. Age was strongly associated with mortality with a difference of medians (DoM) of 13.15 years (95% confidence interval (CI) 11.37 to 14.94) between those who died and those who survived. We found a clinically relevant difference between non-survivors and survivors for C-reactive protein (CRP; DoM 69.10 mg/L, CI 50.43 to 87.77), lactate dehydrogenase (LDH; DoM 189.49 U/L, CI 155.00 to 223.98), cardiac troponin I (cTnI; DoM 21.88 pg/mL, CI 9.78 to 33.99) and D-Dimer (DoM 1.29mg/L, CI 0.9 to 1.69). Furthermore, cerebrovascular disease was the co-morbidity most strongly associated with mortality (Odds Ratio 3.45, CI 2.42 to 4.91) and ICU admission (Odds Ratio 5.88, CI 2.35 to 14.73).

Discussion

This comprehensive meta-analysis found age, cerebrovascular disease, CRP, LDH and cTnI to be the most important risk-factors that predict severe COVID-19 outcomes and will inform clinical scores to support early decision-making.

Introduction

Coronavirus disease (COVID-19) was declared a pandemic by the World Health Organization (WHO) on March 11th, 2020 [1]. As of October 31st 2020 approximately 46 million people were infected with this virus [2]. The outcomes of COVID-19 vary from completely asymptomatic to hospitalization, ICU admission and death [3, 4].

Several studies aimed to identify possible risk factors for a severe outcome. Studies investigated demographic risk factors and found advanced age to be the strongest predictor of a severe course [57]. However, age alone does not explain the variability in the severity of disease with sufficient granularity [8]. Symptoms on presentation associated with severe disease include dyspnea, fever, cough, and fatigue [6, 9, 10]. Several co-morbidities have been identified as risk factors, including cardiovascular disease, obesity, chronic respiratory disease, diabetes, cerebrovascular disease, chronic renal failure and cancer [7, 1118]. The effect of other co-morbidities on disease outcome remain less clear: e.g. hypertension being associated with a decreased risk [7, 19] for death in some and an increased risk [20] in other publications. Similarly, data on past and current smoking are inconsistent in respect to the association with disease severity [2125]. Biomarkers predicting severe disease include different markers of inflammation and acute phase reaction (e.g. CRP, procalcitonin (PCT), white blood cells (WBC), lymphopenia, interleukin 6 (IL-6)) [26, 27]. Increased D-dimer levels, as a marker for coagulation and thrombosis, were found to be elevated in non-survivors, whereas other coagulation markers failed to show statistical and clinical difference [13, 2830]. Markers indicating cardiac damage, such as cardiac troponin I or T and N terminal pro B type natriuretic peptide (NT-proBNP) were also associated with severe disease and mortality [31].

This systematic review aims, to our knowledge for the first time, to comprehensively evaluate demographic, clinical and laboratory indicators for their association with severe COVID-19 and death.

Methods

This trial was registered at PROSPERO on April 4th, 2020 (Registration number: CRD42020177154). The PRISMA checklist is provided in the supplementary S2 in S1 File.

Eligibility criteria

Studies eligible for inclusion provided data on demographic, clinical and/or laboratory risk factors for the following outcomes: hospitalization, intubation, ICU admission, and/or death. Laboratory values and vital parameters taken at hospital admission were considered. Cross-sectional studies, cohort studies, randomized and non-randomized controlled trials were included. No specific restrictions were placed in terms of demographic and clinical characteristics of the population being studied. The search was conducted on July 29th, the search date was set from December 1st 2019 to May 31st 2020.

A full list of data items screened for in the studies is available in the supplementary S12 in S1 File. These data items were chosen, on the one hand, according to the information available in the existing literature and, on the other hand, in order to identify risk factors at hospital admission.

Search strategy

Medline [PubMed] and Web of Science Core Collection as well as preprint databases (bioRxiv and medRxiv) were searched. The exact search terms were developed with an experienced medical librarian (GG) using combinations of subject headings (when applicable) and text-words for the concepts without language restrictions. The full search strategy used for PubMed is presented in the supplementary S1 in S1 File. The results of the search term were imported into the bibliography manager Zotero (Version 5.0.92) for further processing.

Study screening and data extraction

Study selection was done by three authors (SK, CG and JG) initially in parallel for five randomly selected papers and after alignment in the selection was guaranteed, it was done independently by each of the reviewers. Article title and abstracts were screened for eligibility in English or German language, followed by a full-text review for those eligible.

A structured electronic data extraction form was developed (AS, LMH, SO, LAS and BP), piloted on five randomly selected papers and then used to extract information from included studies. Six reviewers (SK, CG, SS, SaK, MG and AM) performed data extraction in duplicate for the first five randomly selected papers to ensure alignment and then independently, with concerns being discussed jointly. For continuous indicators we extracted means and standard deviation as well as medians, first quartiles and third quartiles if available. The comprehensive list of data items that were collected is presented in the supplementary S12 in S1 File. Throughout screening and extraction, disagreements were discussed until consensus was reached, and a senior author (CMD) was consulted when necessary.

Given the concern for reporting of the same patients in different publications [32] leading to a bias in the data, we excluded papers which included patients from the same hospital with an overlapping inclusion date. Furthermore, we excluded data from 23 articles (peer-reviewed and preprint), because the reported laboratory values with the reported units were obviously incorrect (supplementary S3 in S1 File), unless we were able to clarify the issue with the authors of the respective paper directly.

Assessment of study quality

To analyze risk of bias in individual studies, we evaluated the studies using an approach adapted from an existing Cochrane tool by Higgins et al. [33] for systematic reviews that assessed indicators of outcomes. Specifically, we analyzed three areas: 1) case definition and severity definition; 2) patient data availability and exclusions and; 3) selection bias and applicability. We rated the risk of bias in low, intermediate and high risks of bias.

Statistical analysis

We grouped indicators into binary and continuous indicators across five categories: (1) demographics, (2) symptoms, (3) co-morbidities, (4) laboratory and (5) clinical course/treatment. We analyzed all available indicators between (1) hospitalized and non-hospitalized patients, (2) ICU-admitted patients and non-ICU admitted patients, (3) intubated and non-intubated patients, and (4) patients who died and patients who survived. Most data were available for ICU admission and death. Thus, we focus on these comparison groups in the main paper and present data on hospitalization and intubation in the supplement. Clinical significance was determined by expert consensus with clinicians. A predefined rule (e.g. 10% above normal range) across biomarkers or vital parameters is not possible as it is different for every marker and the unit chosen.

Meta-analyses were only performed when there were at least 4 primary studies reporting adequate summary data. As the continuous indicators were often skewed and were summarized by medians in most primary studies, we meta-analyzed the difference of medians across groups for continuous indicators. Specifically, we pooled the difference of medians in a random effects meta-analysis using the Quantile Estimation (QE) approach proposed by McGrath et al. [34]. In secondary analyses, median value of indicators in each comparison group were pooled using the same approach.

The QE approach estimates the variance of the (difference of) medians in studies that report the sample median and first and third quartiles of the outcome. When studies report sample means and standard deviations of the outcome, this approach estimates the (difference of) medians and its variance. Then, the standard inverse-variance approach is applied to obtain a pooled estimate of the population (difference of) medians. The population difference of medians can be interpreted as the difference between the median value of the indicator in one group (e.g., those who survived) and the median value of the indicator in the other group (e.g., those who died).

For binary indicators, the pooled odds ratios (OR) and associated 95% confidence intervals (CI) were estimated in a random effects meta-analysis. For both binary and continuous indicators, the restricted maximum likelihood (REML) approach was used to estimate between-study heterogeneity. When REML failed to converge for a continuous indicator, we used the DerSimmonian and Laird (DL) estimator for all analyses involving this indicator.

For all analyses, between-study heterogeneity was assessed by the I2 statistic. The presence of small-study effects was visually assessed in funnel plots. Analyses were performed in R (version 4.0.2) with package ‘metamedian’ [35] and in Stata (Version 16.1). The code is publicly available on GitHub (https://github.com/stmcg/covid-ma).

Results

The search resulted in 6,702 articles, of which 3,733 were excluded because they did not present primary data (e.g. guidelines, recommendations, letter to the editors or correspondences, study protocols, modeling), 792 were case reports, 465 focused on patients younger than 18 years and 381 were systematic reviews. In total, 88 articles were included (Fig 1). The majority of studies (52) were conducted in China, 21 in Europe, 12 in the USA, two in Iran, one in South Korea. Most studies were retrospective cohorts (n = 84) and four had a prospective study design. All studies were in English. Data on mortality were reported in 64 studies, data on ICU admission were available in 26 studies (two studies reported both and patients were counted twice). In total, data from 69,762 patients were meta-analyzed, of whom 5,311 died and 57,321 survived and 2,112 provided data on ICU admission while 5,018 did not require ICU admission. We were not able to perform a meta-analysis for all indicators (supplementary S12 in S1 File) extracted from the publications. Meta-analysis for all eligible indicators for each outcome is listed in the supplementary section (S5–S8 in S1 File).

Study quality

The findings on study quality can be found in Fig 2. When considering the case and severity definition of COVID-19, almost 50% of studies were considered low risk of bias, while only 9.1% had a high risk. In contrast, many studies were identified to have high concerns for bias in respect to patient selection and generalizability of findings (36.4% high risk, 9.1% low risk). In more than a third of studies, we had high concern that the full data on patients were not available and inappropriate exclusion might have occurred (35.2%). The full explanation of the risk of bias assessment and the assessment of each paper individually is available in the supplement S4 in S1 File. Overall, high- or intermediate risk of bias for at least one category was found in almost three fourth (73.8%) of studies. No study scored low risk in all three categories.

ICU admission

Fig 3 and Table 1 show the pooled odds ratios (OR) and differences of medians (DoM), respectively, for ICU admission for the different indicators in the five categories: demographic, symptoms, comorbidities, laboratory and clinical values [12, 13, 29, 3657].

thumbnail
Fig 3. Pooled odds ratios among ICU vs. non ICU groups.

ICU = Intensive care unit, OR = odds ratio, CI = confidence interval, COPD = chronic obstructive pulmonary disease, ART = anti-retroviral treatment, NIV = non-invasive ventilation.

https://doi.org/10.1371/journal.pone.0255154.g003

thumbnail
Table 1. Summary of the meta-analysis results for continuous indicators comparing those who were admitted to the ICU and those who were not.

https://doi.org/10.1371/journal.pone.0255154.t001

Patients requiring ICU admission had a median age of 65 years (CI 62.27 to 66.16). Those not requiring ICU admission were significantly younger with a median age of 59 years (CI 55.93 to 61.86) with a DoM of 4.63 years (CI 1.43 to 7.82) (Table 1). We were not able to perform a subgroup analysis of different age groups as data provided by primary studies was insufficient.

Of the many possible symptoms of COVID-19, we found dyspnea (OR 5.34, CI 2.77 to 10.28) and fatigue (OR 1.63, CI 1.20 to 2.22) to be significantly associated with ICU admission. In terms of co-morbidities, patients admitted to the ICU were more likely to suffer from cerebrovascular disease (OR 5.88, CI 2.35 to 14.73), hypertension (OR 1.62 CI 1.24 to 2.12), diabetes (OR 1.58, CI 1.29 to 1.93) and chronic kidney disease (OR 1.48, CI 1.08 to 2.03). In contrast, cardiovascular diseases (OR 1.50, CI 0.99 to 2.28), chronic obstructive pulmonary disease (COPD) (OR 1.39, CI 0.90 to 2.16), chronic lung disease (OR 1.06, CI 0.89 to 1.25) and smoking (OR 1.00, CI 0.77 to 1.29) were not associated with ICU admission.

Few laboratory values showed differences between patients that required ICU admission and those who did not (Table 1). D-dimer failed to show a statistically significant difference (DoM 0.3 mg/L, CI -0.2 to 0.81). We found a clinically relevant elevation of CRP and cardiac Troponin I (cTnI) in patients requiring ICU admission, although cTnI failed to be statistically significant (DoM for CRP 56.41 mg/L, CI 39.8 to 73.02 and DoM for cTnI 19.27 pg/mL, CI -4.13 to 42.68). A clinically significant reduction in lymphocytes was also observed (DoM -0.34, CI -0.39 to -0.29). Leukocytes, neutrophiles and LDH were also significantly higher in patients admitted to an ICU, but the absolute elevation over those in non-ICU patients were small and of questionable clinical relevance (Table 1).

Patients developing acute kidney failure, as a complication at any stage, had the highest risk for ICU admission (OR 15.69, CI 11.22 to 21.90).

Mortality

Fig 4 and Table 2 show the pooled odds ratios and differences of medians, respectively, for mortality for symptoms, comorbidities, laboratory and clinical values [11, 16, 28, 58110].

thumbnail
Fig 4. Pooled odds ratios among mortality vs. survived groups.

OR = odds ratio, CI = confidence interval, COPD = chronic obstructive pulmonary disease, ART = anti-retroviral treatment, ECMO = extracorporeal membrane oxygenation, NIV = non-invasive ventilation.

https://doi.org/10.1371/journal.pone.0255154.g004

thumbnail
Table 2. Summary of the meta-analysis results for continuous indicators comparing those who died and those who survived.

https://doi.org/10.1371/journal.pone.0255154.t002

Patients who died had a median age of 71 years (CI 69.3 to 71.61) compared to survivors with a median age of 58 years (CI 55.03 to 59.4) for a DoM of 13.15 years (CI 11.37 to 14.94] (Table 2). Again, dyspnea was the symptom that differentiated markedly between survivors and non-survivors (OR 3.69, CI 2.54 to 5.36). Also, fatigue was more frequently observed in those who died (OR 1.48, CI 1.15 to 1.89). Regarding vital parameters at admission, patients who died presented with a median peripheral oxygen saturation (SpO2) on room air of 89% (CI 87.32 to 90.91) to the hospital, while those who survived had 95% (CI 94.59 to 96.63) (DoM -6.33%, CI -8.14 to -4.52).

Patients who died were more likely to suffer from cardiovascular disease (OR 3.93, CI 2.91 to 5.30), cerebrovascular disease (OR 3.45, CI 2.42 to 4.91), chronic lung disease (OR 3.12, CI 2.17 to 4.49), COPD (OR 2.54, CI 1.87 to 3.44; Fig 4) and hypertension (OR 2.49, CI 2.11 to 2.94). Current and former smokers had an increased risk of mortality (OR 1.36, CI 1.10 to 1.67). Patients with chronic kidney disease (CKD) (OR 2.36, CI 1.89 to 2.94), diabetes (OR 2.14, CI 1.82 to 2.52) and cancer (OR 2.08, CI 1.55 to 2.77) also had an increased odds of mortality. Co-morbidities not associated with increased odds of mortality were asthma, liver disease, digestive system disease and immunosuppressive therapy (Fig 4). Clinically relevant elevations outside the normal laboratory range in patients who died compared to those who survived were observed in two markers of inflammation: CRP was elevated by 69.1mg/L (CI 50.43 to 87.77) and IL-6 by 31.19 pg/mL (CI 11.96 to 50.41). Furthermore, clinically significant elevations were observed in cTnI by 21.88pg/mL (CI 9.78 to 33.99) and D-dimer by 1.29mg/L (CI 0.9 to 1.69), while lymphocytes were significantly lower: -0.34x109/L (CI -0.39 to -0.29). Other makers (hemoglobin, leukocytes, neutrophils, platelets, international normalized ratio (INR), Prothrombin, alanine transaminase (ALAT), aspartate transaminase (ASAT), Albumin, LDH, blood urea nitrogen (BUN), Creatinine, PCT, BNP, CK and creatine kinase myocardial band (CK-MB)) were also significantly elevated in those who died. However, the absolute difference compared to those who survived was small and thus likely not clinically relevant. For leukocytes, neutrophils, platelets, prothrombin, ALAT, ASAT, BUN, Creatinine, CK and CK-MB the point estimates even stayed within the normal laboratory range.

As a clinical complication, acute kidney injury showed the highest overall odds ratio for mortality (OR 20.87, CI 9.21 to 47.32), followed by requiring non-invasive ventilation (NIV) (OR 7.38, CI 4.25 to 12.82).

Fig 5 shows pooled median estimates along with their normal laboratory ranges for selected number of indicators among patients who died, patients who survived, ICU-admitted patients, and non-ICU admitted patients. Pooled difference of medians estimates for all indicators are available in the supplementary files (S5 for mortality in S1 File, S6 for ICU admission in S1 File, S7 for intubation and hospitalization in S8 in S1 File). After removing large outliers in a sensitivity analyses for CRP and D-dimer, results did not change substantially (results available in the supplementary S9 in S1 File). Funnel plots showed no substantial asymmetry suggesting small-study effects except for data assessing acute kidney injury (supplementary S10 for mortality and S11 for ICU admission in S1 File).

thumbnail
Fig 5. Pooled median estimates of selected indicators along with their normal laboratory ranges among patients who died, patients who survived, ICU-admitted patients, and non-ICU admitted patients.

ICU = Intensive care unit, CRP = C-reactive protein, LDH = Lactate dehydrogenase.

https://doi.org/10.1371/journal.pone.0255154.g005

Discussion

In this comprehensive systematic review and meta-analysis, we corroborate known markers of severe disease for COVID-19 and shed light on further indicators, whose significance was indeterminate to date.

With respect to co-morbidities, we identified cardiovascular disease, which includes chronic heart disease and coronary artery disease (OR 3.93), chronic lung disease (OR 3.12) and COPD (OR 2.54) as strong risk factors of mortality among COVID-19 patients but not for ICU admission. Only cerebrovascular disease was strongly associated with an increased risk of both ICU admission and death (almost six- and three-fold higher ratio for ICU admission and death, respectively). Overall, the finding that cerebrovascular disease is associated with poor outcomes is in line with the more recent data highlighting the importance of delirium and an overall depressed mental state in severe COVID-19 [111113]. Our findings found chronic kidney disease, diabetes [18] and COPD/chronic lung disease to be risk factors, however, associations are less strong than those for cardiovascular or cerebrovascular disease [111]. Evidence from previous studies regarding the risk associated with hypertension has been inconclusive. Our work identifies hypertension as a clear risk factor for ICU admission (OR 1.62, CI 1.24 to 2.12) and death (OR 2.49, CI 2.11 to 2.94) [7, 19, 20]. Similarly, while prior data were inconclusive with respect to the influence of smoking for severe COVID-19 [3, 2225], our meta-analysis shows the increased risk of mortality among smokers (OR 1.36, CI 1.10 to 1.67). However, our data did not allow for meta-regression to assess whether this effect was independent of the risk associated with chronic lung disease. In line with some recent studies on asthma, we could not find an increased risk for mortality [114] in our meta-analysis (OR 0.88, CI 0.58 to 1.35).

CRP was the only laboratory marker that was associated with a higher risk of ICU admission (DoM 56.41 mg/L) and death (DoM 69.1 mg/L), while D-dimer elevation was only significantly associated with death (DoM 1.29 mg/L), but not with ICU admission. Although the median elevation of cTnI was clinically relevant both in those who were admitted to the ICU (DoM 19.27 pg/mL) and those who died (DoM 21.88 pg/mL), only in those who died was the difference statistically significant.

We were able to identify clinically relevant lymphopenia as a marker. Lymphopenia was a marker that was used early on for triage purposes to predict disease severity [115] and our findings on increased odds for mortality corroborate the systematic review results on this topic published by Huang and Pranata [116].

For categorical variables, OR was used to measure the association between the outcome (mortality, ICU admission, hospitalization, intubation) and the risk factor/biomarker of interest. We decided to use the OR instead of the relative risk (RR) given the nature of the research question and the data that were available to us. For example, calculating the risk of ICU among people with fever compared to the risk of ICU among people without a fever is not as informative as computing the odds ratio of ICU admission between fever and non-fever patients. We acknowledge, however, when interpreting the results that the ORs are more extreme (further from the null) than the RRs whenever there is a non-null association.

In line with previous reviews, we acknowledge risk factors such as age, dyspnea, smoking, diabetes, hypertension and cardiovascular disease to be associated with increased odds for mortality.

On the one hand we could not find an increased odds for asthma, whereas ‘respiratory diseases’ [3] had an increased OR in other reviews. On the other hand, we find COPD to be a strong risk factor for mortality. This can be explained by different data extractions and pooling of similar diseases. Nevertheless, this suggests that lung diseases are associated with an increased risk of a severe course.

Strengths and limitations of this study

Our study provides a comprehensive review of the data from both pre-print and peer-reviewed sources with a broad geographic distribution and assesses the different categories of risk factors from symptoms, co-morbidities and laboratory values to clinical complications. Correlating the indicators to the two clinical outcomes death and ICU admission, has both strengths and limitations. While ICU admission is a clinical decision, it is, especially early on in a new disease, sometimes a measure of precaution. This might weaken the association of indicators with clinical outcomes. At the same time, when capacity of ICU beds is exhausted, triage decisions might have been made based on age and co-morbidities to not admit to the ICU, thus strengthening an association of an indicator beyond what would be expected under routine conditions. We also assessed the association with hospitalization and intubation (see supplement), but here confounding factors seemed to be even more pronounced, and data are further limited. Confounding factors that lead to this conclusion are for example different health care systems across the globe with or without the possibility of self-care in less severe cases instead of hospital admission or the change in the approach regarding early intubation from the first wave towards a more conservative approach with novel methods such as ‘awake proning’.

In addition, with improving care and novel therapies certain associations might be less pronounced. We did not observe improved survival with antiviral therapy in the studies included, suggesting that this effect might not yet have occurred in the time frame of studies included here. We have not assessed radiological findings as these would likely correlate with clinical signs and symptoms, as well as changes in laboratory parameters. In contrast to other systematic reviews, we did not focus on the course of the disease (e.g. critical, severe), but rather on the outcome [3]. Furthermore risk factors were assessed for each outcome individually [17].

Additional limitations primarily relate to data quality of the included studies. Our quality assessment of studies clearly indicated that substantial bias was present across studies. Primarily the selection bias as suggested for example by the high case fatality rate (e.g. Zhou et al. [11], 28.3%, Chen et al. [117] 11.1%, and Huang et al. [12] 14.6%) is likely to have impacted our results and prospective data collection to confirm findings of these studies is important [118]. However, an analysis of quality was performed at the study level. Conceivably a high-quality study will contribute more high quality data on the risk-factor outcome association, but this cannot be ascertained. In addition, 13 studies were still in preprint at the time of extraction. Furthermore, we found a large number of studies (n = 21, list available in supplementary S3 in S1 File) to include laboratory values that were clinically out of range, which suggests that despite peer-review in some of them, the rush of publication in this pandemic impacted the quality of reporting [119].

Conclusion

Our data on mortality and ICU admission corroborate most of the proposed indicators of clinical outcomes, clarifies the strength of association and highlights additional indicators. In addition, this systematic review highlights the limitations of the studies published and calls for better quality in prospective collections.

Supporting information

S1 File. Supplementary file with supporting information.

This contains supporting information for all outcomes with summary forest plots for ‘intubation’ and ‘hospitalization’, risk of bias assessment and funnel plots.

https://doi.org/10.1371/journal.pone.0255154.s002

(DOCX)

Acknowledgments

We thank Genevieve Gore for her help with the search terms. We thank Sinan Onogur, Laura Aguilera Saiz, and Bünyamin Pekdemir for their help setting up the data extraction tool.

References

  1. 1. World Health Organization (WHO). Coronavirus disease 2019 (COVID-19)Situation Report—72. 2020 27.10.2020]; Available from: https://www.who.int/docs/default-source/coronaviruse/situation-reports/20200401-sitrep-72-covid-19.pdf?sfvrsn=3dd8971b_2.
  2. 2. Worldometers.info. COVID-19 Coronavirus Pandemic. 2020 02.11.2020]; Available from: https://www.worldometers.info/coronavirus/.
  3. 3. Zheng Z., et al., Risk factors of critical & mortal COVID-19 cases: A systematic literature review and meta-analysis. J Infect, 2020. 81(2): p. e16–e25. pmid:32335169
  4. 4. Chen R., et al., Risk Factors of Fatal Outcome in Hospitalized Subjects With Coronavirus Disease 2019 From a Nationwide Analysis in China. Chest, 2020. 158(1): p. 97–105. pmid:32304772
  5. 5. Xie Y., et al., Epidemiologic, clinical, and laboratory findings of the COVID-19 in the current pandemic: systematic review and meta-analysis. BMC Infect Dis, 2020. 20(1): p. 640. pmid:32867706
  6. 6. Li J., et al., Epidemiology of COVID-19: A systematic review and meta-analysis of clinical characteristics, risk factors, and outcomes. J Med Virol, 2020.
  7. 7. Williamson E.J., et al., Factors associated with COVID-19-related death using OpenSAFELY. Nature, 2020. 584(7821): p. 430–436. pmid:32640463
  8. 8. Romero Starke K., et al., The Age-Related Risk of Severe Outcomes Due to COVID-19 Infection: A Rapid Review, Meta-Analysis, and Meta-Regression. Int J Environ Res Public Health, 2020. 17(16).
  9. 9. Arentz M., et al., Characteristics and Outcomes of 21 Critically Ill Patients With COVID-19 in Washington State. JAMA, 2020. pmid:32191259
  10. 10. Tang J.W., et al., Comparing hospitalised, community and staff COVID-19 infection rates during the early phase of the evolving COVID-19 epidemic. J Infect, 2020. 81(4): p. 647–679. pmid:32442455
  11. 11. Zhou F., 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): p. 1054–1062. pmid:32171076
  12. 12. Huang C., et al., Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet, 2020. 395(10223): p. 497–506. pmid:31986264
  13. 13. Wang D., et al., Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China. JAMA, 2020. 323(11): p. 1061–1069. pmid:32031570
  14. 14. Fang L., Karakiulakis G., and Roth M., Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection? The Lancet Respiratory Medicine, 2020. 8(4): p. e21. pmid:32171062
  15. 15. Guan W.-j., et al., Clinical Characteristics of Coronavirus Disease 2019 in China. New England Journal of Medicine, 2020. 382(18): p. 1708–1720.
  16. 16. Yang X., et al., Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med, 2020. 8(5): p. 475–481. pmid:32105632
  17. 17. Rod J.E., Oviedo-Trespalacios O., and Cortes-Ramirez J., A brief-review of the risk factors for covid-19 severity. Rev Saude Publica, 2020. 54: p. 60. pmid:32491116
  18. 18. Nandy K., et al., Coronavirus disease (COVID-19): A systematic review and meta-analysis to evaluate the impact of various comorbidities on serious events. Diabetes Metab Syndr, 2020. 14(5): p. 1017–1025. pmid:32634716
  19. 19. Drager L.F., et al., Is Hypertension a Real Risk Factor for Poor Prognosis in the COVID-19 Pandemic? Curr Hypertens Rep, 2020. 22(6): p. 43. pmid:32535705
  20. 20. Gao C., et al., Association of hypertension and antihypertensive treatment with COVID-19 mortality: a retrospective observational study. Eur Heart J, 2020. 41(22): p. 2058–2066. pmid:32498076
  21. 21. Williamson E.J., et al., OpenSAFELY: factors associated with COVID-19 death in 17 million patients. Nature, 2020.
  22. 22. Zhao Q., et al., The impact of COPD and smoking history on the severity of COVID-19: A systemic review and meta-analysis. J Med Virol, 2020. pmid:32293753
  23. 23. Guo F.R., Active smoking is associated with severity of coronavirus disease 2019 (COVID-19): An update of a meta-analysis. Tob Induc Dis, 2020. 18: p. 37. pmid:32382258
  24. 24. Vardavas C.I. and Nikitara K., COVID-19 and smoking: A systematic review of the evidence. Tobacco Induced Diseases, 2020. 18(March).
  25. 25. World Health Organization (WHO). Smoking and COVID-19. 2020 18.10.2020]; Available from: https://www.who.int/news-room/commentaries/detail/smoking-and-covid-19.
  26. 26. Herold T., et al., Elevated levels of IL-6 and CRP predict the need for mechanical ventilation in COVID-19. J Allergy Clin Immunol, 2020. 146(1): p. 128–136 e4. pmid:32425269
  27. 27. Lippi G. and Plebani M., Procalcitonin in patients with severe coronavirus disease 2019 (COVID-19): A meta-analysis. Clin Chim Acta, 2020. 505: p. 190–191. pmid:32145275
  28. 28. Tang N., et al., Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost, 2020. 18(4): p. 844–847. pmid:32073213
  29. 29. Wang F., et al., Clinical Characteristics of 28 Patients with Diabetes and Covid-19 in Wuhan, China. Endocr Pract, 2020. 26(6): p. 668–674. pmid:32357072
  30. 30. Cummings M.J., et al., Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 in New York City: a prospective cohort study. Lancet, 2020. 395(10239): p. 1763–1770. pmid:32442528
  31. 31. Gao L., et al., Prognostic value of NT-proBNP in patients with severe COVID-19. Respir Res, 2020. 21(1): p. 83. pmid:32293449
  32. 32. Bauchner H., Golub R.M., and Zylke J., Editorial Concern-Possible Reporting of the Same Patients With COVID-19 in Different Reports. JAMA, 2020. 323(13): p. 1256. pmid:32176775
  33. 33. Higgins JPT, S.J., Page MJ, Elbers RG, Sterne JAC, Chapter 8: Assessing risk of bias in a randomized trial, in Cochrane Handbook for Systematic Reviews of Interventions version 6.1 (updated September 2020), T.J. Higgins JPT, Chandler J, Cumpston M, Li T, Page MJ, Welch VA, Editor. 2020.
  34. 34. McGrath S., et al., Meta-analysis of the difference of medians. Biom J, 2020. 62(1): p. 69–98. pmid:31553488
  35. 35. McGrath, S.S., Russell; Benedett, Andrea metamedian: Meta-Analysis of Medians. R package version 0.1.5. 2020; Available from: https://cran.r-project.org/web/packages/metamedian/metamedian.pdf.
  36. 36. Urra J.M., et al., Selective CD8 cell reduction by SARS-CoV-2 is associated with a worse prognosis and systemic inflammation in COVID-19 patients. Clin Immunol, 2020. 217: p. 108486. pmid:32479985
  37. 37. Martin-Moro F., et al., Survival study of hospitalised patients with concurrent COVID-19 and haematological malignancies. Br J Haematol, 2020. 190(1): p. e16–e20. pmid:32379921
  38. 38. Sun S., et al., Abnormalities of peripheral blood system in patients with COVID-19 in Wenzhou, China. Clin Chim Acta, 2020. 507: p. 174–180. pmid:32339487
  39. 39. Hong K.S., et al., Clinical Features and Outcomes of 98 Patients Hospitalized with SARS-CoV-2 Infection in Daegu, South Korea: A Brief Descriptive Study. Yonsei Med J, 2020. 61(5): p. 431–437. pmid:32390367
  40. 40. Lei S., et al., Clinical characteristics and outcomes of patients undergoing surgeries during the incubation period of COVID-19 infection. EClinicalMedicine, 2020. 21: p. 100331. pmid:32292899
  41. 41. Cholankeril G., et al., Association of Digestive Symptoms and Hospitalization in Patients With SARS-CoV-2 Infection. Am J Gastroenterol, 2020. 115(7): p. 1129–1132. pmid:32618665
  42. 42. Antinori S., et al., Compassionate remdesivir treatment of severe Covid-19 pneumonia in intensive care unit (ICU) and Non-ICU patients: Clinical outcome and differences in post-treatment hospitalisation status. Pharmacol Res, 2020. 158: p. 104899. pmid:32407959
  43. 43. Cao M., et al., Clinical Features of Patients Infected with the 2019 Novel Coronavirus (COVID-19) in Shanghai, China. medRxiv, 2020.
  44. 44. Smadja D.M., et al., Angiopoietin-2 as a marker of endothelial activation is a good predictor factor for intensive care unit admission of COVID-19 patients. Angiogenesis, 2020. 23(4): p. 611–620. pmid:32458111
  45. 45. Ferguson J., et al., Characteristics and Outcomes of Coronavirus Disease Patients under Nonsurge Conditions, Northern California, USA, March-April 2020. Emerg Infect Dis, 2020. 26(8): p. 1679–1685. pmid:32407284
  46. 46. Israelsen S.B., et al., Characteristics of patients with COVID-19 pneumonia at Hvidovre Hospital, March-April 2020. Dan Med J, 2020. 67(6).
  47. 47. Yang L., et al., Epidemiological and clinical features of 200 hospitalized patients with corona virus disease 2019 outside Wuhan, China: A descriptive study. J Clin Virol, 2020. 129: p. 104475. pmid:32485619
  48. 48. Criel M., et al., Venous thromboembolism in SARS-CoV-2 patients: only a problem in ventilated ICU patients, or is there more to it? Eur Respir J, 2020. 56(1).
  49. 49. Rieg S., et al., [COVID-19-Response—Strategies of the Task-Force Coronavirus and experiences upon implementation in the management of 115 cases at the University Medical Center Freiburg]. Dtsch Med Wochenschr, 2020. 145(10): p. 657–664. pmid:32344439
  50. 50. Zhang G., et al., Clinical features and short-term outcomes of 221 patients with COVID-19 in Wuhan, China. J Clin Virol, 2020. 127: p. 104364. pmid:32311650
  51. 51. Memtsoudis S.G., et al., Obesity as a risk factor for poor outcome in COVID-19-induced lung injury: the potential role of undiagnosed obstructive sleep apnoea. Br J Anaesth, 2020. 125(2): p. e262–e263. pmid:32439072
  52. 52. Lodigiani C., et al., Venous and arterial thromboembolic complications in COVID-19 patients admitted to an academic hospital in Milan, Italy. Thromb Res, 2020. 191: p. 9–14. pmid:32353746
  53. 53. Middeldorp S., et al., Incidence of venous thromboembolism in hospitalized patients with COVID-19. J Thromb Haemost, 2020. 18(8): p. 1995–2002. pmid:32369666
  54. 54. Myers L.C., et al., Characteristics of Hospitalized Adults With COVID-19 in an Integrated Health Care System in California. JAMA, 2020. 323(21): p. 2195–2198. pmid:32329797
  55. 55. Rentsch C.T., et al., Covid-19 Testing, Hospital Admission, and Intensive Care Among 2,026,227 United States Veterans Aged 54–75 Years. medRxiv, 2020.
  56. 56. Argenziano M.G., et al., Characterization and clinical course of 1000 patients with coronavirus disease 2019 in New York: retrospective case series. BMJ, 2020. 369: p. m1996. pmid:32471884
  57. 57. Petrilli C.M., et al., Factors associated with hospital admission and critical illness among 5279 people with coronavirus disease 2019 in New York City: prospective cohort study. BMJ, 2020. 369: p. m1966. pmid:32444366
  58. 58. Li K., et al., Radiographic Findings and other Predictors in Adults with Covid-19. medRxiv, 2020: p. 2020.03.23.20041673.
  59. 59. Zhang F., et al., Myocardial injury is associated with in-hospital mortality of confirmed or suspected COVID-19 in Wuhan, China: A single center retrospective cohort study. medRxiv, 2020: p. 2020.03.21.20040121.
  60. 60. Fu L., et al., Influence factors of death risk among COVID-19 patients in Wuhan, China: a hospital-based case-cohort study. medRxiv, 2020: p. 2020.03.13.20035329.
  61. 61. Wang Z., et al., Elevated serum IgM levels indicate poor outcome in patients with coronavirus disease 2019 pneumonia: A retrospective case-control study. medRxiv, 2020: p. 2020.03.22.20041285.
  62. 62. Xie J., et al., Development and external validation of a prognostic multivariable model on admission for hospitalized patients with COVID-19. medRxiv, 2020: p. 2020.03.28.20045997.
  63. 63. Russo V., et al., Clinical impact of pre-admission antithrombotic therapy in hospitalized patients with COVID-19: A multicenter observational study. Pharmacol Res, 2020. 159: p. 104965. pmid:32474087
  64. 64. Shi Q., et al., Clinical Characteristics and Risk Factors for Mortality of COVID-19 Patients With Diabetes in Wuhan, China: A Two-Center, Retrospective Study. Diabetes Care, 2020. 43(7): p. 1382–1391. pmid:32409504
  65. 65. Raoufi M., et al., Correlation between Chest Computed Tomography Scan Findings and Mortality of COVID-19 Cases; a Cross sectional Study. Arch Acad Emerg Med, 2020. 8(1): p. e57. pmid:32613199
  66. 66. Luo X., et al., Prognostic value of C-reactive protein in patients with COVID-19. Clin Infect Dis, 2020. pmid:32344321
  67. 67. Tang N., et al., Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost, 2020. 18(5): p. 1094–1099. pmid:32220112
  68. 68. Wang L., et al., Coronavirus disease 2019 in elderly patients: Characteristics and prognostic factors based on 4-week follow-up. J Infect, 2020. 80(6): p. 639–645. pmid:32240670
  69. 69. Conversano A., et al., Renin-Angiotensin-Aldosterone System Inhibitors and Outcome in Patients With SARS-CoV-2 Pneumonia: A Case Series Study. Hypertension, 2020. 76(2): p. e10–e12. pmid:32383626
  70. 70. Cao J., et al., Clinical Features and Short-term Outcomes of 102 Patients with Coronavirus Disease 2019 in Wuhan, China. Clin Infect Dis, 2020. 71(15): p. 748–755. pmid:32239127
  71. 71. Yang X., et al., Extracorporeal Membrane Oxygenation for Coronavirus Disease 2019-Induced Acute Respiratory Distress Syndrome: A Multicenter Descriptive Study. Crit Care Med, 2020. 48(9): p. 1289–1295. pmid:32427613
  72. 72. Giacomelli A., et al., 30-day mortality in patients hospitalized with COVID-19 during the first wave of the Italian epidemic: A prospective cohort study. Pharmacol Res, 2020. 158: p. 104931. pmid:32446978
  73. 73. Goicoechea M., et al., COVID-19: clinical course and outcomes of 36 hemodialysis patients in Spain. Kidney Int, 2020. 98(1): p. 27–34. pmid:32437770
  74. 74. Borghesi A., et al., Chest X-ray severity index as a predictor of in-hospital mortality in coronavirus disease 2019: A study of 302 patients from Italy. Int J Infect Dis, 2020. 96: p. 291–293. pmid:32437939
  75. 75. Zhang F., et al., Obesity predisposes to the risk of higher mortality in young COVID-19 patients. J Med Virol, 2020. pmid:32437016
  76. 76. Wu C., et al., Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease 2019 Pneumonia in Wuhan, China. JAMA Intern Med, 2020. 180(7): p. 934–943. pmid:32167524
  77. 77. Huang J., et al., Hypoalbuminemia predicts the outcome of COVID-19 independent of age and co-morbidity. J Med Virol, 2020. pmid:32406952
  78. 78. Valeri A.M., et al., Presentation and Outcomes of Patients with ESKD and COVID-19. J Am Soc Nephrol, 2020. 31(7): p. 1409–1415. pmid:32467113
  79. 79. Zhang J., et al., The clinical data from 19 critically ill patients with coronavirus disease 2019: a single-centered, retrospective, observational study. Z Gesundh Wiss, 2020: p. 1–4.
  80. 80. Hu H., Yao N., and Qiu Y., Comparing Rapid Scoring Systems in Mortality Prediction of Critically Ill Patients With Novel Coronavirus Disease. Acad Emerg Med, 2020. 27(6): p. 461–468. pmid:32311790
  81. 81. Lee L.Y., et al., COVID-19 mortality in patients with cancer on chemotherapy or other anticancer treatments: a prospective cohort study. Lancet, 2020. 395(10241): p. 1919–1926. pmid:32473682
  82. 82. Yuan M., et al., Association of radiologic findings with mortality of patients infected with 2019 novel coronavirus in Wuhan, China. PLoS One, 2020. 15(3): p. e0230548. pmid:32191764
  83. 83. Chen T., et al., Clinical characteristics of 113 deceased patients with coronavirus disease 2019: retrospective study. BMJ, 2020. 368: p. m1091. pmid:32217556
  84. 84. Tu W.J., et al., Clinicolaboratory study of 25 fatal cases of COVID-19 in Wuhan. Intensive Care Med, 2020. 46(6): p. 1117–1120. pmid:32253448
  85. 85. Auld S.C., et al., ICU and Ventilator Mortality Among Critically Ill Adults With Coronavirus Disease 2019. Crit Care Med, 2020. 48(9): p. e799–e804. pmid:32452888
  86. 86. Sun H., et al., Risk Factors for Mortality in 244 Older Adults With COVID-19 in Wuhan, China: A Retrospective Study. J Am Geriatr Soc, 2020. 68(6): p. E19–E23. pmid:32383809
  87. 87. Yang K., et al., Clinical characteristics, outcomes, and risk factors for mortality in patients with cancer and COVID-19 in Hubei, China: a multicentre, retrospective, cohort study. Lancet Oncol, 2020. 21(7): p. 904–913. pmid:32479787
  88. 88. Chen R., et al., Longitudinal hematologic and immunologic variations associated with the progression of COVID-19 patients in China. J Allergy Clin Immunol, 2020. 146(1): p. 89–100. pmid:32407836
  89. 89. Zhao X., et al., Early decrease in blood platelet count is associated with poor prognosis in COVID-19 patients-indications for predictive, preventive, and personalized medical approach. EPMA J, 2020: p. 1–7.
  90. 90. Deng G., et al., Clinical determinants for fatality of 44,672 patients with COVID-19. Crit Care, 2020. 24(1): p. 179. pmid:32345311
  91. 91. Zou X., et al., Acute Physiology and Chronic Health Evaluation II Score as a Predictor of Hospital Mortality in Patients of Coronavirus Disease 2019. Crit Care Med, 2020. pmid:32697506
  92. 92. Li L., et al., Association of clinical and radiographic findings with the outcomes of 93 patients with COVID-19 in Wuhan, China. Theranostics, 2020. 10(14): p. 6113–6121. pmid:32483442
  93. 93. Fan H., et al., Cardiac injuries in patients with coronavirus disease 2019: Not to be ignored. Int J Infect Dis, 2020. 96: p. 294–297. pmid:32437935
  94. 94. Crespo M., et al., COVID-19 in elderly kidney transplant recipients. Am J Transplant, 2020. 20(10): p. 2883–2889. pmid:32471001
  95. 95. Nowak B., et al., Clinical characteristics and short-term outcomes of patients with coronavirus disease 2019: a retrospective single-center experience of a designated hospital in Poland. Pol Arch Intern Med, 2020. 130(5): p. 407–411. pmid:32420710
  96. 96. Klang E., et al., Severe Obesity as an Independent Risk Factor for COVID-19 Mortality in Hospitalized Patients Younger than 50. Obesity (Silver Spring), 2020. 28(9): p. 1595–1599. pmid:32445512
  97. 97. Pan F., et al., Factors associated with death outcome in patients with severe coronavirus disease-19 (COVID-19): a case-control study. Int J Med Sci, 2020. 17(9): p. 1281–1292. pmid:32547323
  98. 98. Yan X., et al., Neutrophil to lymphocyte ratio as prognostic and predictive factor in patients with coronavirus disease 2019: A retrospective cross-sectional study. J Med Virol, 2020. pmid:32458459
  99. 99. Xu P.P., et al., Risk factors for adverse clinical outcomes with COVID-19 in China: a multicenter, retrospective, observational study. Theranostics, 2020. 10(14): p. 6372–6383. pmid:32483458
  100. 100. Moon A.M., et al., High mortality rates for SARS-CoV-2 infection in patients with pre-existing chronic liver disease and cirrhosis: Preliminary results from an international registry. J Hepatol, 2020. 73(3): p. 705–708. pmid:32446714
  101. 101. Wang K., et al., Clinical and laboratory predictors of in-hospital mortality in patients with COVID-19: a cohort study in Wuhan, China. Clin Infect Dis, 2020.
  102. 102. Paranjpe I., et al., Clinical Characteristics of Hospitalized Covid-19 Patients in New York City. medRxiv, 2020.
  103. 103. Yu C., et al., Clinical Characteristics, Associated Factors, and Predicting COVID-19 Mortality Risk: A Retrospective Study in Wuhan, China. Am J Prev Med, 2020. 59(2): p. 168–175. pmid:32564974
  104. 104. Tomlins J., et al., Clinical features of 95 sequential hospitalised patients with novel coronavirus 2019 disease (COVID-19), the first UK cohort. J Infect, 2020. 81(2): p. e59–e61. pmid:32353384
  105. 105. Javanian M., et al., Clinical and laboratory findings from patients with COVID-19 pneumonia in Babol North of Iran: a retrospective cohort study. Rom J Intern Med, 2020. 58(3): p. 161–167. pmid:32396143
  106. 106. Qi X., et al., Clinical course and risk factors for mortality of COVID-19 patients with pre-existing cirrhosis: a multicentre cohort study. Gut, 2020. pmid:32434831
  107. 107. Xu B., et al., Suppressed T cell-mediated immunity in patients with COVID-19: A clinical retrospective study in Wuhan, China. J Infect, 2020. 81(1): p. e51–e60. pmid:32315725
  108. 108. Deng Y., et al., Clinical characteristics of fatal and recovered cases of coronavirus disease 2019 in Wuhan, China: a retrospective study. Chin Med J (Engl), 2020. 133(11): p. 1261–1267. pmid:32209890
  109. 109. Wang D., et al., Clinical course and outcome of 107 patients infected with the novel coronavirus, SARS-CoV-2, discharged from two hospitals in Wuhan, China. Crit Care, 2020. 24(1): p. 188. pmid:32354360
  110. 110. Du R.H., et al., Predictors of mortality for patients with COVID-19 pneumonia caused by SARS-CoV-2: a prospective cohort study. Eur Respir J, 2020. 55(5).
  111. 111. Atkins J.L., et al., Preexisting Comorbidities Predicting COVID-19 and Mortality in the UK Biobank Community Cohort. J Gerontol A Biol Sci Med Sci, 2020. 75(11): p. 2224–2230. pmid:32687551
  112. 112. Mao L., et al., Neurologic Manifestations of Hospitalized Patients With Coronavirus Disease 2019 in Wuhan, China. JAMA Neurol, 2020. 77(6): p. 683–690. pmid:32275288
  113. 113. Pleasure S.J., Green A.J., and Josephson S.A., The Spectrum of Neurologic Disease in the Severe Acute Respiratory Syndrome Coronavirus 2 Pandemic Infection: Neurologists Move to the Frontlines. JAMA Neurol, 2020. 77(6): p. 679–680. pmid:32275291
  114. 114. Schultze A., et al., Risk of COVID-19-related death among patients with chronic obstructive pulmonary disease or asthma prescribed inhaled corticosteroids: an observational cohort study using the OpenSAFELY platform. Lancet Respir Med, 2020. pmid:32979987
  115. 115. Tan L., et al., Lymphopenia predicts disease severity of COVID-19: a descriptive and predictive study. Signal Transduct Target Ther, 2020. 5(1): p. 33. pmid:32296069
  116. 116. Huang I. and Pranata R., Lymphopenia in severe coronavirus disease-2019 (COVID-19): systematic review and meta-analysis. J Intensive Care, 2020. 8: p. 36. pmid:32483488
  117. 117. Chen N., et al., Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet, 2020. 395(10223): p. 507–513. pmid:32007143
  118. 118. Jakob C.E.M., et al., First results of the "Lean European Open Survey on SARS-CoV-2-Infected Patients (LEOSS)". Infection, 2020. pmid:33001409
  119. 119. Bramstedt K.A., The carnage of substandard research during the COVID-19 pandemic: a call for quality. J Med Ethics, 2020.