COVID-19 outcomes among hospitalized men with or without exposure to alpha-1-adrenergic receptor blocking agents ================================================================================================================ * Shilong Li * Tomi Jun * Zichen Wang * Yu-Han Kao * Emilio Schadt * Maximilian F. Konig * Chetan Bettegowda * Joshua T. Vogelstein * Nickolas Papadopoulos * Ramon E. Parsons * Rong Chen * Eric E. Schadt * Li Li * William K. Oh ## Abstract **Importance** Alpha-1-adrenergic receptor antagonists (α1-blockers) can abrogate pro-inflammatory cytokines and may improve outcomes among patients with respiratory infections. Repurposing readily available drugs such as α1-blockers could augment the medical response to the COVID-19 pandemic. **Objective** To evaluate the association between α1-blocker exposure and COVID-19 mortality **Design** Real-world evidence study **Setting** Patient level data with 32,355 records tested for SARS-CoV-2 at the Mount Sinai Health System including 8,442 laboratory-confirmed cases extracted from five member hospitals in the New York City metropolitan area. **Participants** 2,627 men aged 45 or older admitted with COVID-19 between February 24 and May 31, 2020 **Exposures** α1-blocker use as an outpatient or while admitted for COVID-19 **Main Outcomes and Measures** In-hospital mortality **Results** Men exposed to α1-blockers (N=436) were older (median age 73 vs. 64 years, P<0.001) and more likely to have comorbidities than unexposed men (N=2,191). Overall, 758 (28.9%) patients died in hospital, 1,589 (60.5%) were discharged, and 280 (10.7%) were still hospitalized as of May 31, 2020. Outpatient exposure to α1-blockers was not associated with COVID-19 hospital outcomes, though there was a trend towards significance (OR 0.749, 95% CI 0.527-1.064; P=0.106). Conversely, inpatient use of α1-blockers was independently associated with improved in-hospital mortality in both multivariable logistic (OR 0.633, 95% CI 0.434-0.921; P=0.017) and Cox regression analyses (HR 0.721, 95% CI 0.572-0.908; P=0.006) adjusting for patient demographics, comorbidities, and baseline vitals and labs. Age-stratified analyses suggested greater benefit from inpatient α1-blocker use among younger age groups: Age 45-65 OR 0.384, 95% CI 0.164-0.896 (P=0.027); Age 55-75 OR 0.511, 95% CI 0.297-0.880 (P=0.015); Age 65-89 OR 0.810, 95% CI 0.509-1.289 (P=0.374). **Conclusions and Relevance** Inpatient α1-blocker use was independently associated with improved COVID-19 mortality among hospitalized men. Clinical trials to assess the therapeutic value of α1-blockers in COVID-19 are warranted. ## Introduction Severe coronavirus disease 2019 (COVID-19) has been linked to dysregulated immune responses, including an overexuberant inflammatory response marked by high levels of proinflammatory cytokines such as interleukin-6 (IL-6).1,2 Immunosuppressive drugs such as glucocorticoids have become a standard component of treatment for severe COVID-19,3 and several trials of anti-cytokine or anti-inflammatory agents are underway or have reported promising results.4–10 Despite these advances, there remains a pressing need for safe, effective, and widely available therapeutic options. Adrenergic signaling has been linked to hyperinflammation in models of bacterial sepsis and cytokine release syndrome. In pre-clinical experiments, a positive feedback loop of adrenergic signaling was identified wherein macrophages responded to catecholamines by producing more catecholamines and inflammatory cytokines; this adrenergic loop could be interrupted by blockade of α1-adrenergic receptors with prazosin.11 In a retrospective clinical study of patients with acute respiratory distress and pneumonia, exposure to α1-adrenergic receptor antagonists (α1-blockers) was associated with a significant reduction in risk of mechanical ventilation or death.12 Similarly, a recent retrospective analysis of 25,130 patients with COVID-19 across the United States Veterans Health Administration hospital system showed that outpatient exposure to any α1-blocker was associated with decreased in-hospital mortality compared to matched controls not on any α1-blocker at the time of hospital admission.13 These observations have led to the hypothesis that α1-blockers in routine clinical use (e.g. prazosin, doxazosin, tamsulosin, etc.) may be repurposed for COVID-19 treatment.14 We therefore conducted this real-world evidence study based on electronic medical record (EMR) data to determine whether exposure to α1-blockers is independently associated with mortality among patients hospitalized with COVID-19. ## Methods ### Data sources This retrospective study utilized de-identified electronic medical record (EMR; Epic systems, Verona, WI) data from 5 member hospitals within the Mount Sinai Health System (MSHS) in the New York City metropolitan area. De-identified EMR data were obtained via the Mount Sinai Data Warehouse ([https://labs.icahn.mssm.edu/msdw/](https://labs.icahn.mssm.edu/msdw/)). We identified 8,442 MSHS patients with PCR confirmed diagnosis of COVID-19 from February 24 through May 31, 2020 during the peak of pandemic in NYC. COVID-19 was diagnosed by real-time reverse transcriptase polymerase chain reaction (RT-PCR)-based clinical tests from nasopharyngeal swab specimens. We retrieved patient demographics, social history, medication history, and disease comorbidities from the EMR including age, gender, race/ethnicity, smoking status, asthma, chronic obstructive pulmonary disease (COPD), hypertension, obstructive sleep apnea, obesity, diabetes, chronic kidney disease, human immunodeficient virus (HIV) infection, cancer, coronary artery disease, atrial fibrillation, heart failure, chronic viral hepatitis, alcoholic nonalcoholic liver disease, and acute kidney injury (AKI). Patients aged ≥ 89 years were assigned an age of 89 to prevent re-identification. Medications by prescription or hospital administration captured in EMR from January 1, 2019 till May 31, 2020 were included in the medication history. We identified disease comorbidities through their corresponding ICD-10-CM codes before hospital admission and during hospitalization. We also extracted data from each hospital encounter, including vital sign and laboratory data at the time of presentation, and medications administered during hospitalization. Vital sign and laboratory data extracted included: white blood cell count (WBC), serum creatinine, anion gap, potassium, alanine aminotransferase (ALT), body mass index (BMI), temperature, oxygen saturation, heart rate, respiratory rate, systolic blood pressure (SBP) and diastolic blood pressure (DBP). We defined three possible outcomes for each hospitalization: in-hospital death (deceased), discharged to home or other locations not associated with acute medical care (recovered), and continued hospitalization (right censored). The duration of hospitalization was calculated from the beginning of the hospital encounter till death or discharge. This study was approved by the Mount Sinai institutional review board (IRB): IRB-17-01245. ### Study design This was a retrospective EMR-based study designed to test the independent association of α1-blocker exposure with in-hospital death among COVID-19 patients. We first identified 6,218 inpatients positive for COVID-19 in one of 5 hospital systems within the MSHS as of May 31, 2020 (Figure 1). The majority (93%) of α1-blocker users in this cohort were men aged 45 or older. We therefore limited the analysis cohort to men aged 45 or older (N=2,627). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/04/11/2021.04.08.21255148/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/F1) Figure 1. Schematic flowchart of data extraction and analysis plan We defined α1-blocker exposure as an active prescription from January 1, 2020 for an α1-blocker (tamsulosin, alfuzosin, silodosin, terazosin, doxazosin, and prazosin) up to and including hospitalization for COVID-19 (N=436). We further defined a subset of patients (N=343) with documented α1-blocker administration during their hospitalization, which we defined as “α1-blocker inpatient use”. Potential confounders in the analysis included demographic characteristics, comorbidities, baseline labs and vitals, and exposure to medications used to treat hypertension, hyperlipidemia, and inflammation. Detailed medication names included in these categories are shown in supplement Table S1. ### Statistical analysis Patient characteristics were summarized as median and interquartile range (IQR) for continuous variables or mean and standard deviation (SD). We displayed categorical variables as number and percentage (%). We performed a statistical test of hypothesis for differences using the Kruskal-Wallis test or two sample t-test for continuous variables, and the χ2 test for categorical variables. We employed multivariate logistic regression models with potential confounders to estimate the odds ratio and corresponding 95% confidence interval for COVID-19 in-hospital mortality (deceased=1) versus recovery (recovered=0) associated with α1-blocker use. We considered the following potential confounders: age, hospital stay duration, race, smoking status, BMI, temperature, O2 saturation, heart rate, respiratory rate, hypertension, asthma, COPD, obstructive sleep apnea, obesity, diabetes mellitus, chronic kidney disease, HIV, cancer, coronary artery disease, atrial fibrillation, heart failure, chronic viral hepatitis, liver disease, AKI, ICU stay, WBC, creatinine, anion gap, potassium, and ALT. We applied the Wald-based power and sample size formulas for logistic regression to calculate the power given the observed sample size.15 We used two-tailed test to estimate the probability of event under the null hypothesis which was the in-hospital mortality rate and we used the binomial distribution for α1-blocker with the prescription rate as the probability. To account for right-censored patients (N=280), we used multivariate Cox proportional hazard models to evaluate the association between α1-blockers and survival (in-hospital mortality). We estimated the hazard ratio of α1-blockers on COVID-19 patient in-hospital survival, adjusting for the same confounders mentioned above. We utilized propensity score weighting to adjust the potential confounders between those exposed or unexposed to α1-blockers. We utilized the R package ‘twang’ and calculated the propensity scores based on average treatment effect on the treated (ATT) and Kolmogorov-Smirnov evaluation criterion. We then employed the ‘survey’ R package by fitting logistic regression models with propensity scores as the weights to assess the α1-blocker treatment effect on COVID-19 patients. Statistical significance was defined as a two-sided P-value < 0.05, unless otherwise noted. ### Sensitivity analysis To evaluate the robustness of the associations between α1-blockers and COVID-19 in-hospital mortality, we performed sensitivity analysis to assess the impact from potential unmeasured or uncontrolled confounding. We calculated E-values for odds and hazard ratios as point estimates, in addition to E-values for their upper confidence limits.16 The point estimates represent the strength of unmeasured or uncontrolled confounding required to explain away the observed associations. The upper limits signify the strength of unmeasured or uncontrolled confounding needed to move the confidence interval to include the null, i.e. OR=1 or HR=1. If the effect of unmeasured or uncontrolled confounding is weaker than the calculated E-value, the association of α1-blockers with COVID-19 in-hospital mortality could not be fully biased by a weaker confounder. ## Results ### Patient characteristics and outcomes We identified 6,218 inpatients positive for COVID-19 admitted to one of 5 hospitals within the MSHS as of May 31, 2020. Among these 6,218 patients, 464 patients (7.5%) had been prescribed α1-blockers between January 1, 2020 up to and including their hospitalization, with at least 2 months prior to admission; most of these patients were men aged 45 or older (N=436, 93%). Therefore, we limited the analysis cohort to men aged 45 years or older (N=2,627). This cohort was divided into an α1-blocker-exposed group (N=436) and an unexposed group (N=2,191). All patients in the α1-blocker group had been prescribed α1-blockers at some point from January 1, 2020 up to and including their COVID-19 hospitalization (“α1-blocker exposure”); a subset of these patients (N=343) had documented α1-blocker administration during their hospitalization (“inpatient α1-blocker use”). The α1-blocker exposed group was older (median age 73 vs. 64 years, P<0.001) and more likely to have comorbidities than the unexposed group. Chronic diseases such as COPD, hypertension, diabetes mellitus, chronic kidney disease, cancer, and cardiovascular disease were significantly enriched in the α1-blocker group. Additional patient demographics for the two groups are shown in Table 1 and Figure S1. View this table: [Table 1.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T1) Table 1. Baseline characteristics and hospitalization outcomes, by alpha-1-blocker exposure Overall, 758 (28.9%) patients died, 1,589 (60.5%) were discharged, and 280 (10.7%) were still hospitalized as of May 31, 2020. Unadjusted in-hospital mortality was 32.3% in the exposed group and 28.2% in the unexposed group. ### α1-blocker exposure and COVID-19 in-hospital mortality We evaluated the association of α1-blockers on COVID-19 patients with known outcomes (death: N=758; discharge: N=1,589) using multivariate logistic-regression models. In the overall population, α1-blocker exposure was not significantly associated with in-hospital mortality (OR 0.749; 95% CI, 0.527-1.064; P=0.106) with the power β = 0.62 based on the sample size of 2347. There was a trend towards a protective association for α1-blocker exposure in the subset of patients who were hospitalized for at least 24 hours, though this was not significant (N=2,276; OR 0.722; 95% CI, 0.505-1.034; P=0.075) (Table 2). This might be due to limited sample size (β = 0.71 in the Wald-based power analysis). View this table: [Table 2.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T2) Table 2. Multivariable logistic regression results for COVID-19 in-hospital mortality, by cohort (overall vs. hospitalization >1 day) and alpha-1-blocker exposure (any vs. inpatient use) To account for right-censored patients (N=280), we used Cox proportional hazard models to evaluate the association of α1-blocker exposure and COVID-19 in-hospital survival. There was a trend towards a protective effect of α1-blocker exposure in the overall cohort (HR 0.808; 95% CI, 0.652-1.002; P=0.052) and among those hospitalized for at least 24 hours (HR 0.823; 95% CI, 0.660-1.027; P=0.085), though these associations were not significant (Table 3). View this table: [Table 3.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T3) Table 3. Multivariable Cox regression results for COVID-19 in-hospital mortality, by cohort (overall vs. hospitalization >1 day) and alpha-1-blocker exposure (any vs. inpatient use) ### Inpatient α1-blocker use and COVID-19 in-hospital mortality We further assessed the impact of α1-blockers on mortality for patients with documented administration of α1-blockers while admitted to the hospital (N=343) compared to unexposed patients. We observed that inpatient α1-blocker use significantly reduced the risk of in-hospital mortality overall (OR 0.633; 95% CI 0.434-0.921; P=0.017) and among those hospitalized for at least 24 hours (OR 0.607; 95% CI, 0.415-0.890; P=0.01) (Table 2). The survival analysis also showed improved in-hospital survival for those with inpatient α1-blocker use overall (HR 0.721; 95% CI, 0.572-0.908; P=0.006), and among patients hospitalized for at least 24 hours (HR 0.751; 95% CI, 0.593-0.950; P=0.017) (Table 3). ### Sensitivity analysis We assessed the impact of potential unmeasured or uncontrolled confounding on the observed associations between inpatient α1-blockers use and COVID-19 in-hospital mortality using the E-value metric (Table S2).16 Based on this analysis, an unmeasured confounder would have to have an odds ratio of at least 1.25 (in the logistic regression analysis) or a hazard ratio of at least 1.34 (in the survival analysis) to cause the 95% confidence interval for the treatment effect of inpatient α1-blockers use to include the null. ### Age-stratified associations of α1-blockers and COVID-19 in-hospital mortality We assessed the interaction between age and inpatient α1-blocker use for overall cohort and patients with hospital stay > 1 day, respectively, and we did not find any significance between the interaction and in-hospital mortality. Therefore, to identify differences in the treatment effect of α1-blockers on different age groups, we segmented the population into three age groups (45-65; 55-75; 65-89) and analyzed each group separately using logistic regression, adjusting for the same covariates as the unstratified analysis. The age groups were overlapped by 10 years to preserve sample size. Inpatient α1-blocker use was associated with a significantly lower risk of in-hospital mortality in the 45-65 (OR 0.384; 95% CI 0.164-0.896; P=0.027) and 55-75 age groups (OR 0.511; 95% CI 0.297-0.880; P=0.015), but not the 65-89 age group (OR 0.810; 95% CI 0.509-1.289; P=0.374) (Table 4). View this table: [Table 4.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T4) Table 4. Age-stratified multivariable logistic regression and propensity-weighted logistic regression analyses for COVID-19 in-hospital mortality, by cohort (overall vs. hospitalization >1 day) and alpha-1-blocker exposure (any vs. inpatient use) ### Propensity score analysis We further assessed the effect of α1-blockers on in-hospital mortality using propensity-weighted logistic regression. These yielded similar results as the multivariable regression and age-stratified analyses. Inpatient α1-blocker use was associated with reduced in-hospital mortality (OR 0.630; 95% CI 0.424-0.935; P=0.022) whereas α1-blocker exposure was not (OR 0.740; 95% CI 0.512-1.070; P=0.110) (Table S3). In age-stratified analyses, patients in younger age groups had greater benefit from α1-blocker exposure and inpatient α1-blocker use than patients in the oldest age group (Table 4). ## Discussion Using a racially and ethnically diverse cohort from New York City comprising 2,627 men aged 45 or older hospitalized COVID-19 patients seen between February 24 and March 31, 2020, we found that inpatient use of α1-blockers was significantly associated with reduced in-hospital mortality, whereas outpatient exposures were not associated with benefit, although there was a trend towards significance. In age stratification analysis, we found that α1-blocker use was more beneficial to younger age groups. Drug repurposing is the process of finding new indications for drugs already in clinical use. The appeal of rapidly validating and deploying an existing drug against a deadly global pandemic is clear, especially if the drug is widely available and affordable. Dexamethasone, now a standard in COVID-19 treatment, is an example of a commonly used drug repurposed for a new indication.3 However, the saga of hydroxychloroquine, which was touted as a cure early in the pandemic but has since proven ineffective, is a cautionary tale.17 The allure of rapid drug repurposing must be balanced against rigorous scientific method. α1-blockers, commonly used to treat benign prostatic hyperplasia and hypertension, have become a target for drug repurposing due to pre-clinical data linking α1-adrenergic signaling to pro-inflammatory cytokines which may contribute to dysregulated immunity and adverse outcomes in COVID-19.1,2,11 These pre-clinical findings have been bolstered by recent retrospective clinical analyses linking α1-blockers with improved outcomes in hospitalized patients with both COVID-19 and non-COVID-19 respiratory infections.12,13 In a large COVID-19 cohort drawn from the US Veterans Health Administration hospital system, outpatient α1-blocker exposure was associated with a relative risk reduction of 18% for in-hospital mortality compared to matched controls.13 Interestingly, the non-selective α1-blocker doxazosin, which inhibits all three α1-adrenergic receptor subtypes (α1A, α1B, α1D), was associated with a greater relative risk reduction (74%) than the uroselective (α1A, α1D) α1-blocker tamsulosin (18%). In the present study, we found that in-hospital use of α1-blockers was consistently and independently associated with reduced in-hospital mortality using both multivariable regression and propensity score-based methods. In age-stratified analyses, we observed that this protective effect was more pronounced in relatively younger age groups. In contrast to the studies by Koenecke et al. and Rose et al., which defined α1-blocker exposure based on outpatient prescriptions only, we were able to use inpatient medication administration records to identify patients treated with α1-blockers during their COVID-19 hospitalization. These results lend additional support the hypothesis that α1-blockers may have a beneficial effect in COVID-19. Our results also include tests of association between other common medication classes and COVID-19 outcomes, including beta blockers, angiotensin-converting enzyme (ACEi) inhibitors, angiotensin II receptor blockers (ARBs), and glucocorticoids. Results pertaining to these other medications should be taken in the context of a selected cohort designed to study α1-blocker use and COVID-19 outcomes. Exposures to these other classes were not ascertained with the same granularity as they were for the primary exposure. That said, it is interesting to note that α1-blocker and beta blocker exposure were associated with opposite COVID-19 outcomes in our cohort. There is evidence to suggest that β-adrenergic signaling can promote an anti-inflammatory M2 phenotype in macrophages, in contrast to the pro-inflammatory effect of α1-adrenergic signaling.11,18 Additional efforts to dissect the interactions between adrenergic signaling and the COVID-19 immune response are warranted. A prior diagnosis of asthma was associated with reduced in-hospital mortality in this analysis. While this observation deserves further scrutiny, it is conceivable that early exposure to inhaled glucocorticoids or β-adrenergic agonists may have contributed to this signal. ### Limitations Our study has several limitations. The cohort did not include women since most α1-blockers were prescribed to men, most likely for benign prostatic hyperplasia. Male sex is a recognized risk factor for adverse COVID-19 outcomes, possibly due to sex-specific differences in immunity.19 Thus, these results may not extrapolate to women. We did not account for different types of α1-blockers, which differentially target the three α1-adrenergic receptor subtypes. Importantly, a causal relationship cannot be definitively established between α1-blockers and improved COVID-19 outcomes in this retrospective study. Several confounders, such as older age and comorbidities were more common in the α1-blocker group. However, these adverse risk factors would be expected to bias the study result towards the null rather than inflate a protective association. Furthermore, our findings are consistent with prior data, and were robust to different methods of adjustment for confounding and sensitivity analysis. The ongoing randomized clinical trial of prazosin against placebo among hospitalized COVID-19 patients ([NCT04365257](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04365257&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom)) will include women and provide more definitive data on the therapeutic value of α1-blockers. Finally, our study is based on a single center EMR and medication via outpatient use cannot be tracked for adherence. Therefore, analyzing in-hospital medication administration is more robust. Furthermore, EMR in MSHS is the one of the largest and most comprehensive EMR systems, representing racial/ethnic diversity in New York City, and EMR implementation is from various data sources, so our findings are supported by large patient cohort and were robust from applying multiple methods of adjustment for confounding. ## Conclusions In conclusion, this retrospective study found a protective association between α1-blockers use and COVID-19 outcomes in a cohort of hospitalized men. These results augment the rationale for studying and repurposing α1-blockers as a COVID-19 therapeutic. We await the results of the ongoing randomized clinical trial. ## Data Availability The Mount Sinai Health System database is not publicly available. We utilized Python and R, and their open-source libraries to conduct our analysis tailored to MSH data. Therefore, we are unable to publicly release the code since it is useless without the dataset available. ## Author contributions conceived and designed the study: WKO, LL and SL; performed data extraction from MSHS: SL, ZW, ES; performed statistical analysis: SL; contributed clinical interpretation: TJ, MFK, CB, JV, NP, EES, RP, RC, LL, WKO; wrote and edited the paper: SL, TJ, MFK, CB, JV, NP, EES, RP, RC, LL, WKO. ## Conflict of interest Sema4 is a for-profit company currently majority owned by the Icahn School of Medicine at Mount Sinai (ISMMS). RC, EES, LL and WKO receive compensation from Sema4 that includes equity in the company. In addition to their roles with Sema4, RC, EES, LL and WKO remain affiliated with ISSMS as part-time employees and faculty members. WKO also has consulted for AAA, Astellas, AstraZeneca, Bayer, Conjupro, Foundry, Janssen, Merck, Sanofi and TeneoBio. The JHU filed a patent application on the use of various drugs to prevent cytokine release syndromes, on which NP, BV, KWK, and SZ are listed as inventors. JHU will not assert patent rights from this filing for treatment related to COVID-19. NP is a founder of, consultant to and holds equity in Thrive an Exact company. NP is a founder of and holds equity in Personal Genome Diagnostics. NP is an advisor to and holds equity in Cage Pharma, ManaTbio and NeoPhore. CB is a consultant to Depuy-Synthes and Bionaut Labs. CB, MFK, BV, KWK, and NP are also inventors on technologies unrelated or indirectly related to the work described in this article. Licenses to these technologies are or will be associated with equity or royalty payments to the inventors, as well as to JHU. The terms of all these arrangements are being managed by JHU in accordance with its conflict of interest policies. ## Data and materials availability The Mount Sinai Health System database is not publicly available. We utilized Python and R, and their open-source libraries to conduct our analysis tailored to MSH data. Therefore, we are unable to publicly release the code since it is useless without the dataset available. ## SUPPLEMENTARY MATERIALS ![Figure S1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/04/11/2021.04.08.21255148/F2.medium.gif) [Figure S1.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/F2) Figure S1. Age distributions of α1-blockers exposed and unexposed groups View this table: [Table S1a.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T5) Table S1a. Medication categories and individual medications View this table: [Table S1b.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T6) Table S1b. Number of patients exposed to glucocorticoid medications View this table: [Table S2.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T7) Table S2. Sensitivity analysis using E-values to assess the threshold effect size of unmeasured confounders required to nullify the significant associations between inpatient alpha-1-blocker use and COVID-19 in-hospital mortality View this table: [Table S3.](http://medrxiv.org/content/early/2021/04/11/2021.04.08.21255148/T8) Table S3. Inverse propensity-weighted logistic regression for COVID-19 in-hospital mortality, by cohort (overall vs. hospitalization >1 day) and alpha-1-blocker exposure (any vs. inpatient use) ## Acknowledgement The authors would like to acknowledge Sema4 IT for computational support hosted on AWS. JV is supported by grants from Microsoft Research and FastGrants. * Received April 8, 2021. * Revision received April 8, 2021. * Accepted April 10, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Qin C, Zhou L, Hu Z, et al. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clinical Infectious Diseases. 2020;71(15):762–768. doi:10.1093/cid/ciaa248 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/ciaa248&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32161940&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom) 2. 2.Del Valle DM, Kim-Schulze S, Huang H-H, et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nature Medicine. 2020;26(10):1636–1643. doi:10.1038/s41591-020-1051-9 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-1051-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32839624&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom) 3. 3.The RECOVERY Collaborative Group. Dexamethasone in Hospitalized Patients with Covid-19 — Preliminary Report. New England Journal of Medicine. 2020;0(0):null. doi:10.1056/NEJMoa2021436 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2021436&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32678530&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom) 4. 4.Salama C, Han J, Yau L, et al. Tocilizumab in Patients Hospitalized with Covid-19 Pneumonia. New England Journal of Medicine. 2021;384(1):20–30. doi:10.1056/NEJMoa2030340 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2030340&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33332779&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom) 5. 5.Hermine O, Mariette X, Tharaux P-L, et al. Effect of Tocilizumab vs Usual Care in Adults Hospitalized With COVID-19 and Moderate or Severe Pneumonia: A Randomized Clinical Trial. JAMA Intern Med. 2021;181(1):32. doi:10.1001/jamainternmed.2020.6820 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jamainternmed.2020.6820&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33080017&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom) 6. 6.The REMAP-CAP Investigators, Gordon AC, Mouncey PR, et al. Interleukin-6 Receptor Antagonists in Critically Ill Patients with Covid-19 – Preliminary report. medRxiv. Published online January 7, 2021:2021.01.07.21249390. doi:10.1101/2021.01.07.21249390 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wMS4wNy4yMTI0OTM5MHYyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMTEvMjAyMS4wNC4wOC4yMTI1NTE0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. 7.RECOVERY Collaborative Group, Horby PW, Pessoa-Amorim G, et al. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): preliminary results of a randomised, controlled, open-label, platform trial. medRxiv. Published online February 11, 2021:2021.02.11.21249258. doi:10.1101/2021.02.11.21249258 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wMi4xMS4yMTI0OTI1OHYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMTEvMjAyMS4wNC4wOC4yMTI1NTE0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 8. 8.Benjamin D. Randomized Master Protocol for Immune Modulators for Treating COVID-19. clinicaltrials.gov; 2021. Accessed February 15, 2021. [https://clinicaltrials.gov/ct2/show/NCT04593940](https://clinicaltrials.gov/ct2/show/NCT04593940) 9. 9.Tardif J-C, Bouabdallaoui N, L’Allier PL, et al. Efficacy of Colchicine in Non-Hospitalized Patients with COVID-19. medRxiv. Published online January 27, 2021:2021.01.26.21250494. doi:10.1101/2021.01.26.21250494 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMS4wMS4yNi4yMTI1MDQ5NHYxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMTEvMjAyMS4wNC4wOC4yMTI1NTE0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 10. 10.Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus Remdesivir for Hospitalized Adults with Covid-19. New England Journal of Medicine. 2020;0(0):null. doi:10.1056/NEJMoa2031994 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2031994&link_type=DOI) 11. 11.Staedtke V, Bai R-Y, Kim K, et al. Disruption of a self-amplifying catecholamine loop reduces cytokine release syndrome. Nature. 2018;564(7735):273–277. doi:10.1038/s41586-018-0774-y [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-018-0774-y&link_type=DOI) 12. 12.Koenecke A, Powell M, Xiong R, et al. Alpha-1 adrenergic receptor antagonists to prevent hyperinflammation and death from lower respiratory tract infection. arXiv:arxiv:200410117 [q-bio]. Published online September 8, 2020. Accessed February 16, 2021. [http://arxiv.org/abs/2004.10117](http://arxiv.org/abs/2004.10117) 13. 13.Rose L, Graham L, Koenecke A, et al. The Association Between Alpha-1 Adrenergic Receptor Antagonists and In-Hospital Mortality from COVID-19. medRxiv. Published online February 11, 2021:2020.12.18.20248346. doi:10.1101/2020.12.18.20248346 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoibWVkcnhpdiI7czo1OiJyZXNpZCI7czoyMToiMjAyMC4xMi4xOC4yMDI0ODM0NnYyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDQvMTEvMjAyMS4wNC4wOC4yMTI1NTE0OC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 14. 14.Konig MF, Powell M, Staedtke V, et al. Preventing cytokine storm syndrome in COVID-19 using α-1 adrenergic receptor antagonists. J Clin Invest. 2020;130(7):3345–3347. doi:10.1172/JCI139642 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI139642&link_type=DOI) 15. 15.Demidenko E. Sample size determination for logistic regression revisited. Stat Med. 2007;26(18):3385–3397. doi:10.1002/sim.2771 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/sim.2771&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17149799&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000248549000002&link_type=ISI) 16. 16.VanderWeele TJ, Ding P. Sensitivity Analysis in Observational Research: Introducing the E-Value. Ann Intern Med. 2017;167(4):268–274. doi:10.7326/M16-2607 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7326/M16-2607&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28693043&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F04%2F11%2F2021.04.08.21255148.atom) 17. 17.Singh B, Ryan H, Kredo T, Chaplin M, Fletcher T. Chloroquine or hydroxychloroquine for prevention and treatment of COVID-19. Cochrane Database of Systematic Reviews. 2021;(2). doi:10.1002/14651858.CD013587.pub2 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/14651858.CD013587.pub2&link_type=DOI) 18. 18.Lamkin DM, Ho H-Y, Ong TH, et al. β-adrenergic-stimulated macrophages: Comprehensive localization in the M1–M2 spectrum. Brain Behav Immun. 2016;57:338–346. doi:10.1016/j.bbi.2016.07.162 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.bbi.2016.07.162&link_type=DOI) 19. 19.Takahashi T, Iwasaki A. Sex differences in immune responses. Science. 2021;371(6527):347–348. doi:10.1126/science.abe7199 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzNzEvNjUyNy8zNDciO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wNC8xMS8yMDIxLjA0LjA4LjIxMjU1MTQ4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==)