torstai 18. kesäkuuta 2020

Blood type A is associated with a 45% increased risk for COVID-19-induced respiratory failure

Blood type A is associated with a 45% increased risk for COVID-19-induced respiratory failure than other blood groups, according to a genome-wide association study in @NEJM

___


June 17, 2020

COVID-19: Blood Type / Mavrilimumab / Prone Position

By Kelly Young


See some of the most recent research developments in COVID-19:

Blood type and risk: A genome-wide association study in the New England Journal of Medicine finds that patients with blood type A are at increased risk for COVID-19-induced respiratory failure than other blood groups.

Researchers compared the genomes of roughly 1600 people with severe COVID-19 in Italy and Spain with over 2200 uninfected population-based controls. Two chromosomal loci were associated with COVID-19-induced respiratory failure. One of these is also the ABO blood group locus.

Blood group A was associated with a 45% increased risk for COVID-19 respiratory failure, while blood group O was associated with a 35% lower risk, relative to other blood groups. The other affected locus covered genes that have functions that could be relevant to severe COVID-19, such as interacting with the SARS-CoV-2 cell-surface receptor.

MavrilimumabThe monoclonal antibody mavrilimumab, an investigational treatment that's been studied for rheumatoid arthritis, is associated with improved clinical outcomes among patients with severe COVID-19 in Italy, suggests a small study in The Lancet Rheumatology.

Thirteen non-mechanically ventilated patients with systemic hyperinflammation who received an intravenous dose of mavrilimumab were compared with 26 patients given usual care. Mortality at 28 days was lower in the mavrilimumab group (0% vs. 27%), but the difference was not statistically significant. The mean time to clinical improvement (i.e., improving two points on a seven-point scale) was significantly faster in the treatment group (8 vs. 19 days). The authors acknowledge that the results need to be confirmed in a large, randomized, placebo-controlled trial.

Prone positioning: Another study has found improved oxygenation in awake, nonintubated patients with COVID-19 after lying in the prone position. In JAMA Internal Medicine, researchers instructed 25 hospitalized COVID-19 patients who were receiving supplemental oxygen and had an oxyhemoglobin saturation (SpO2) of 93% or less to lie on their stomachs for as long as they could each day.

One hour after proning, SpO2 levels increased modestly by a median of 7%. Nineteen of the 25 patients had an SpO2 of at least 95% after 1 hour.

Intubation rates were lower among patients who achieved an SpO2 of at least 95% (37% vs. 83%).

Commentators note: "The optimal timing of intubation and mechanical ventilation for patients with [acute respiratory distress syndrome] is not known, but delayed intubation has been associated with increased mortality in patients with ARDS."



__


Genomewide Association Study of Severe Covid-19 with Respiratory Failure

List of authors.
  • David Ellinghaus, Ph.D., 
  • Frauke Degenhardt, M.Sc., 
  • Luis Bujanda, M.D., Ph.D., 
  • Maria Buti, M.D., Ph.D., 
  • Agustín Albillos, M.D., Ph.D., 
  • Pietro Invernizzi, M.D., Ph.D., 
  • Javier Fernández, M.D., Ph.D., 
  • Daniele Prati, M.D., 
  • Guido Baselli, Ph.D., 
  • Rosanna Asselta, Ph.D., 
  • Marit M. Grimsrud, M.D., 
  • Chiara Milani, Ph.D., 
  •  for The Severe Covid-19 GWAS Group*

Abstract

BACKGROUND

There is considerable variation in disease behavior among patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (Covid-19). Genomewide association analysis may allow for the identification of potential genetic factors involved in the development of Covid-19.

METHODS

We conducted a genomewide association study involving 1980 patients with Covid-19 and severe disease (defined as respiratory failure) at seven hospitals in the Italian and Spanish epicenters of the SARS-CoV-2 pandemic in Europe. After quality control and the exclusion of population outliers, 835 patients and 1255 control participants from Italy and 775 patients and 950 control participants from Spain were included in the final analysis. In total, we analyzed 8,582,968 single-nucleotide polymorphisms and conducted a meta-analysis of the two case–control panels.

RESULTS

We detected cross-replicating associations with rs11385942 at locus 3p21.31 and with rs657152 at locus 9q34.2, which were significant at the genomewide level (P<5×10−8) in the meta-analysis of the two case–control panels (odds ratio, 1.77; 95% confidence interval [CI], 1.48 to 2.11; P=1.15×10−10; and odds ratio, 1.32; 95% CI, 1.20 to 1.47; P=4.95×10−8, respectively). At locus 3p21.31, the association signal spanned the genes SLC6A20LZTFL1CCR9FYCO1CXCR6 and XCR1. The association signal at locus 9q34.2 coincided with the ABO blood group locus; in this cohort, a blood-group–specific analysis showed a higher risk in blood group A than in other blood groups (odds ratio, 1.45; 95% CI, 1.20 to 1.75; P=1.48×10−4) and a protective effect in blood group O as compared with other blood groups (odds ratio, 0.65; 95% CI, 0.53 to 0.79; P=1.06×10−5).

CONCLUSIONS

We identified a 3p21.31 gene cluster as a genetic susceptibility locus in patients with Covid-19 with respiratory failure and confirmed a potential involvement of the ABO blood-group system. (Funded by Stein Erik Hagen and others.)
Figure 1.Timeline of Rapid Covid-19 Genomewide Association Study (GWAS).
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was discovered in Wuhan, China, in late 2019, and coronavirus disease 2019 (Covid-19), the disease caused by SARS-CoV-2, rapidly evolved into a global pandemic.1 As of June 15, 2020, there were more than 8.03 million confirmed cases worldwide, with total deaths exceeding 436,900.2 In Europe, Italy and Spain were severely affected early on, with epidemic peaks starting in the second half of February 2020 (Figure 1) and 61,507 deaths reported by June 15, 2020. Covid-19 has varied manifestations,3 with the large majority of infected persons having only mild symptoms or even no symptoms.4 Mortality rates are driven predominantly by the subgroup of patients who have severe respiratory failure related to interstitial pneumonia in both lungs and acute respiratory distress syndrome.5 Severe Covid-19 with respiratory failure requires early and prolonged support by mechanical ventilation.6
The pathogenesis of severe Covid-19 and the associated respiratory failure is poorly understood, but higher mortality is consistently associated with older age and male sex.7,8 Clinical associations have also been reported for hypertension, diabetes, and other obesity-related and cardiovascular disease traits, but the relative role of clinical risk factors in determining the severity of Covid-19 has not yet been clarified.7-11Observational data on lymphocytic endotheliitis and diffuse microvascular and macrovascular thromboembolic complications suggest that Covid-19 is a systemic disease that involves injury to the vascular endothelium but provide little insight into the underlying pathogenesis.12-14 At the peak of the epidemic in Italy and Spain in early 2020, we performed a genomewide association study (GWAS) in an attempt to delineate host genetic factors contributing to severe Covid-19 with respiratory failure. The relatively low disease burden of Covid-19 in Norway and Germany allowed for a complementary team to be set up, whereby genotyping and analysis could occur in parallel with the rapid recruitment of patients in the heavily affected Italian and Spanish epicenters.

Methods

STUDY PARTICIPANTS AND RECRUITMENT

We recruited 1980 patients with severe Covid-19, which was defined as hospitalization with respiratory failure and a confirmed SARS-CoV-2 viral RNA polymerase-chain-reaction (PCR) test from nasopharyngeal swabs or other relevant biologic fluids, cross sectionally, from intensive care units and general wards at seven hospitals in four cities in the pandemic epicenters in Italy and Spain (Table S1A in Supplementary Appendix 1, available with the full text of this article at NEJM.org). The hospitals in Italy were the following: Fondazione IRCCS Cá Granda Ospedale Maggiore Policlinico in Milan (597 patients); Humanitas Clinical and Research Center, IRCCS, in Milan (154 patients); and UNIMIB (Università degli Studi di Milano–Bicocca) School of Medicine, San Gerardo Hospital, in Monza (a suburb of Milan) (200 patients). The hospitals in Spain were the following: Hospital Clínic and IDIBAPS (Instituto de Investigaciones Biomédicas August Pi i Sunyer) in Barcelona (56 patients), Hospital Universitario Vall d’Hebron in Barcelona (337 patients), Hospital Universitario Ramón y Cajal in Madrid (298 patients), and Donostia University Hospital in San Sebastian (338 patients).
Respiratory failure was defined in the simplest possible manner in order to ensure feasibility: the use of oxygen supplementation or mechanical ventilation, with severity graded according to the maximum respiratory support received at any point during hospitalization (supplemental oxygen therapy only, noninvasive ventilatory support, invasive ventilatory support, or extracorporeal membrane oxygenation). For severity assessments, severity was also dichotomized as no mechanical ventilation or mechanical ventilation. Whole-blood samples or buffy coats from diagnostic venipuncture were obtained for DNA extraction.
For comparison, we included 2381 control participants from Italy and Spain (Table S1B in Supplementary Appendix 1). We recruited 998 randomly selected blood donors at Fondazione IRCCS Cá Granda Ospedale Maggiore Policlinico, Milan, who underwent genotyping for the purpose of the present study. A total of 40 of these participants had evidence of the development of anti–SARS-CoV-2 antibodies, all of whom had mild or no Covid-19 symptoms. We also included two control panels with genotype data derived from previous studies and from persons with unknown SARS-CoV-2 infection status using the same genotyping array. The panels included 396 healthy volunteers, blood donors, and outpatients of gastroenterology departments in Italy15 and 987 healthy blood donors in San Sebastian, Spain.

ETHICS COMMITTEE APPROVAL

The project protocol involved the rapid recruitment of patient-participants and no additional project-related procedures (we primarily used material from clinically indicated venipunctures) and afforded anonymity, owing to the minimal data set collected. Differences in recruitment and consent procedures among the centers arose because some centers integrated the project into larger Covid-19 biobanking efforts, whereas other centers did not, and because there were differences in how local ethics committees provided guidance on the handling of anonymization or deidentification of data as well as consent procedures. Written informed consent was obtained, sometimes in a delayed fashion, from the study patients at each center when possible. In some instances, informed consent was provided verbally or by the next of kin, depending on local ethics committee regulations and special policies issued for Covid-19 research. For some severely ill patients, an exemption from informed consent was obtained from a local ethics committee or according to local regulations in order to allow the use of completely anonymized surplus material from diagnostic venipuncture.
The following approvals of the project were obtained from the relevant ethics committees: Germany: Kiel (reference number, D464/20); Italy: Fondazione IRCCS Cá Granda Ospedale Maggiore Policlinico (reference numbers, 342_2020 for patients and 334-2020 for control participants), Humanitas Clinical and Research Center, IRCCS (reference number, 316/20), the University of Milano–Bicocca School of Medicine, San Gerardo Hospital, Monza (the ethics committee of the National Institute of Infectious Diseases Lazzarro Spallanzani reference number, 84/2020); Norway: Regional Committee for Medical and Health Research Ethics in South-Eastern Norway (reference number, 132550); Spain: Hospital Clínic, Barcelona (reference number, HCB/2020/0405), Hospital Universitario Vall d’Hebron, Barcelona (reference number, PR[AG]244/2020), Hospital Universitario Ramón y Cajal, Madrid (reference number, 093/20) and Donostia University Hospital, San Sebastian (reference number, PI2020064).

SAMPLE PROCESSING, GENOTYPING, AND IMPUTATION

We performed DNA extraction using a Chemagic 360 (PerkinElmer) with the use of the low-volume kit CMG-1491 and the buffy-coat kit CMG-714 (Chemagen), respectively. For genotyping, we used the Global Screening Array (GSA), version 2.0 (Illumina), which contains 712,189 variants before quality control. Details on genotyping and quality-control procedures are provided in the Supplementary Methods section in Supplementary Appendix 1. To maximize genetic coverage, we performed single-nucleotide polymorphism (SNP) imputation on genome build GRCh38 using the Michigan Imputation Server and 194,512 haplotypes generated by the Trans-Omics for Precision Medicine (TOPMed) program (freeze 5).16
After the exclusion of samples during quality control (the majority of which were due to population outliers; see the Supplementary Methods section and Table S1B and S1C), the final case–control data sets comprised 835 patients and 1255 control participants from Italy and 775 patients and 950 control participants from Spain. A total of 8,965,091 SNPs were included in the Italian cohort and 9,140,716 SNPs in the Spanish cohort.

STATISTICAL ANALYSIS

To take imputation uncertainty into account, we tested for phenotypic associations with allele dosage data separately for both the Italian and Spanish case–control panels with the use of the PLINK logistic-regression framework for dosage data (PLINK, version 1.9).17 We carried out two genomewide tests of association that included covariates from principal-component analyses, with adjustments to control for potential population stratification (main analysis) and potential population stratification and age and sex bias (analysis corrected for age and sex). A fixed-effects meta-analysis was conducted with the use of the meta-analysis tool METAL18 on 8,582,968 variants that were common to both the Italian and Spanish data sets with the use of effect-size estimates and their standard errors from the study-specific association analyses.
For the genomewide meta-analysis, we used the commonly accepted threshold of 5×10−8for joint P values to determine statistical significance. Bayesian fine-mapping analysis was performed for loci reaching genomewide significance (see the Supplementary Methods section). Genomewide summary statistics of our analyses are publicly available through our web browser (www.c19-genetics.eu. opens in new tab) and have been submitted to the European Bioinformatics Institute (www.ebi.ac.uk/gwas. opens in new tab; accession numbers, GCST90000255. opens in new tab and GCST90000256. opens in new tab).
On the basis of the results from the TOPMed genotype imputation, we selected three ABOSNPs (rs8176747, rs41302905, and rs8176719)19,20 to infer the ABO blood type and calculated odds ratios according to blood type (A vs. B, AB, or O; B vs. A, AB, or O; AB vs. A, B, or O; and O vs. A, AB, or B) (see the Supplementary Methods). To assess in detail the HLA complex at locus 6p21, we performed sequencing-based HLA typing of seven classical HLA loci (HLA-A, -C, -B, -DRB1, -DQA1, -DQB1, and -DPB1) in a subgroup of 835 patients and 891 control participants from Italy and 773 patients from Spain (see the Supplementary Methods). We also assessed allelic distribution according to no mechanical ventilation (supplemental oxygen only) as compared with mechanical ventilation of any type. A similar assessment was made for lead SNPs rs11385942 and rs657152 at loci 3p21.31 and 9q34.2, respectively.

Results

PATIENTS, GENOTYPING, AND QUALITY CONTROL

Table 1.Overview of Patients Included in the Final Analysis.
The milestones of the study in the context of the peak outbreaks in Italy and Spain are shown in Figure 1. Data on the age, sex, maximum respiratory support at any point during hospitalization, and relevant coexisting conditions (type 2 diabetes, hypertension, and coronary heart disease) in the patients who were included in the final analysis are shown in Table 1and in Table S2 in Supplementary Appendix 1. Because we used the same genotyping platform (GSA) to obtain both data sets, we were able to perform a uniform quality control of the merged Italian and Spanish SNP data sets, thus reducing technical confounders to a minimum. A quantile–quantile (Q-Q) plot of the two meta-analyses (the main analysis and the analysis corrected for age and sex) showed significant associations in the tail of the distribution with minimal genomic inflation (λGC=1.015 for main analysis and λGC=1.006 for analysis corrected for age and sex) (Fig. S2 in Supplementary Appendix 1). We also carried out separate association analyses for the Italian and Spanish data sets (see the Supplementary Methods section and Fig. S3).

GENOMEWIDE ASSOCIATION ANALYSIS

Figure 2.GWAS Summary (Manhattan) Plot of the Meta-analysis Association Statistics Highlighting Two Susceptibility Loci with Genomewide Significance for Severe Covid-19 with Respiratory Failure.Table 2.Susceptibility Loci Associated with Severe Covid-19 with Respiratory Failure.
We found two loci to be associated with Covid-19–induced respiratory failure with genomewide significance (P<5×10−8) in the main meta-analysis: the rs11385942 insertion–deletion GA or G variant at locus 3p21.31 (odds ratio for the GA allele, 1.77; 95% confidence interval [CI], 1.48 to 2.11; P=1.15×10−10) and the rs657152 A or C SNP at locus 9q34.2 (odds ratio for the A allele, 1.32; 95% CI, 1.20 to 1.47; P=4.95×10−8) (Figure 2 and Table 2 and Supplementary Appendix 2, available at NEJM.org). Both loci showed nominally significant association in both the Spanish and Italian subanalyses (Table 2). The meta-analysis association results for recessive and heterozygous genetic models for the two meta-analyses (main analysis and the analysis corrected for age and sex) are provided in Supplementary Appendix 3, available at NEJM.org. The imputation quality of the associated markers was good (Table 2 and Supplementary Appendix 2), and manual inspection of genotype cluster plots of genotyped SNPs in these regions showed distinct genotype clouds for homozygous and heterozygous calls (Fig. S4 in Supplementary Appendix 1). Furthermore, the analyses that were corrected for age and sex corroborated the observations at both rs11385942 (meta-analysis odds ratio, 2.11; 95% CI, 1.70 to 2.61; P=9.46×10−12) and rs657152 (meta-analysis odds ratio, 1.39; 95% CI, 1.22 to 1.59; P=5.35×10−7) (Table 2 and Fig. S5 in Supplementary Appendix 1).
Figure 3.Regional Association Plots of Susceptibility Loci Associated with Severe Covid-19 with Respiratory Failure.
The allele frequencies in Spanish and Italian control data sets from previously published studies21-27 are consistent with those we report here (Supplementary Appendix 2). A further 24 different genomic loci showed suggestive evidence (P<1×10−5) for association with Covid-19–induced respiratory failure in the main analysis (Supplementary Appendix 4, available at NEJM.org, and Fig. S6 in Supplementary Appendix 1). Association signals at loci 3p21.31 and 9q34.2 were fine-mapped to 22 and 38 variants, respectively, with greater than 95% certainty (Figure 3A and 3B and Supplementary Appendix 5, available at NEJM.org).

CHROMOSOME 3P21.31

The association signal at locus 3p21.31 comprised six genes (SLC6A20LZTFL1CCR9FYCO1CXCR6, and XCR1) (Figure 3A). The risk allele GA of rs11385942 is associated with reduced expression of CXCR6 and increased expression of SLC6A20, and LZTFL1 is strongly expressed in human lung cells (Fig. S7 and Supplementary Appendix 6, available at NEJM.org). We found that the frequency of the risk allele of the lead variant at 3p21.31 (rs11385942) was higher among patients who received mechanical ventilation than among those who received oxygen supplementation only in both the main meta-analysis (odds ratio, 1.70; 95% CI, 1.27 to 2.26; P=3.30×10−4) and the meta-analysis corrected for age and sex (odds ratio, 1.56; 95% CI, 1.17 to 2.01; P=0.003) (Supplementary Appendix 7, available at NEJM.org). Furthermore, the 19 patients who were homozygous for the rs11385942 risk allele were younger than 1591 patients who were heterozygous or homozygous for the nonrisk allele (median age, 59 years [interquartile range, 49 to 68] vs. 66 years [interquartile range, 56 to 75]; P=0.005). Available variant database entries suggest that the frequency of this risk allele varies among populations worldwide (Fig. S8 in Supplementary Appendix 1).

ABO LOCUS

At locus 9q34.2 the association signal coincided with the ABO blood group locus (Figure 3B and Fig. S9 in Supplementary Appendix 1). Accordingly, the distribution of ABO blood groups (predicted from combinations of genotypes of three different SNPs) was skewed among patients with Covid-19 who had respiratory failure, as compared with the distribution among control participants. In the meta-analysis corrected for age and sex, we found a higher risk among persons with blood group A than among patients with other blood groups (odds ratio, 1.45; 95% CI, 1.20 to 1.75; P=1.48×10−4) and a protective effect for blood group O as compared with the other blood groups (odds ratio, 0.65; 95% CI, 0.53 to 0.79; P=1.06×10−5). Details are provided in Supplementary Appendix 8, available at NEJM.org. Both associations and effect directions were consistent in the separate Spanish and Italian case–control analyses. We found no significant difference in blood-group distribution between patients receiving supplemental oxygen only and those receiving mechanical ventilation of any kind. The ABO blood-group frequency distributions in public registries are provided for comparison in Supplementary Appendix 8, along with details of the results presented here, and corroborate our observations.

HLA ANALYSIS

Given its important role in several viral infections, we scrutinized the extended HLA region (chromosome 6, 25 through 34 Mb). There were no SNP association signals at the HLA complex that met even the significance threshold of suggestive association: P<1×10−5 (Fig. S10 in Supplementary Appendix 1). Dedicated analysis of the classical HLA loci showed no significant allele associations with either Covid-19 or disease severity (oxygen supplementation only or mechanical ventilation of any kind), and further analysis of heterozygote and divergent allele advantage or predicted number of HLA-bound SARS-CoV-2 peptides did not show significant associations with Covid-19 in this data set (see the HLA Analyses section in Supplementary Appendix 1 and Supplementary Appendix 9, available at NEJM.org).

Discussion

Using a pragmatic approach with simplified inclusion criteria and a complementary team of clinicians at the European Covid-19 epicenters in Italy and Spain and scientists in the less-burdened countries of Germany and Norway, we performed a GWAS that included de novo genotyping for Covid-19 with respiratory failure in approximately 2 months. We detected a novel susceptibility locus at a chromosome 3p21.31 gene cluster and confirmed a potential involvement of the ABO blood-group system in Covid-19.
On chromosome 3p21.31, the peak association signal covered a cluster of six genes (SLC6A20LZTFL1CCR9FYCO1CXCR6, and XCR1), several of which have functions that are potentially relevant to Covid-19. A causative gene cannot be reliably implicated by the present data. One candidate is SLC6A20, which encodes the sodium–imino acid (proline) transporter 1 (SIT1) and which functionally interacts with angiotensin-converting enzyme 2, the SARS-CoV-2 cell-surface receptor.28,29 However, the locus also contains genes encoding chemokine receptors, including the CC motif chemokine receptor 9 (CCR9) and the C-X-C motif chemokine receptor 6 (CXCR6), the latter of which regulates the specific location of lung-resident memory CD8 T cells throughout the sustained immune response to airway pathogens, including influenza viruses.30 Flanking genes (e.g., CCR1 and CCR2) also have relevant functions,31 and further studies will be needed to delineate the functional consequences of detected associations.
The preliminary results from the Covid-19 Host Genetics Consortium32 include suggestive associations within the same locus at chromosome 3p21.31, which lend considerable support to our findings (Fig. S11 in Supplementary Appendix 1). The consortium analysis also used population-based controls, but the patients included persons with mild Covid-19 and those with severe Covid-19. The parallel findings nevertheless underscore an important point about the ascertainment of patients and controls in genetic studies of Covid-19. 

Because the majority of patients with SARS-CoV-2 infection are asymptomatic, any sample involving patients with a positive nasopharyngeal RNA test is likely to hold a bias toward some degree of symptomatic burden. Two of the identifiers for inclusion in the current study were a positive result for the presence of SARS-CoV-2 according to PCR testing and receipt of respiratory support (an extreme Covid-19 phenotype). 

As such, it seems reasonable to conclude that the chromosome 3p21.31 locus is involved in Covid-19 susceptibility per se, with a possible enrichment in patients with severe disease. This latter interpretation is supported by the significantly higher frequency of the risk allele among patients who received mechanical ventilation than among those who received supplemental oxygen only as well as by the finding of younger age among patients who were homozygous for the risk allele than among patients who were heterozygous or homozygous for the nonrisk allele.
Nongenetic studies that were reported as preprints33,34 have previously implicated the involvement of ABO blood groups in Covid-19 susceptibility, and ABO blood groups have also been implicated in susceptibility to SARS-CoV-1 infection.35 Our genetic data confirm that blood group O is associated with a risk of acquiring Covid-19 that was lower than that in non-O blood groups, whereas blood group A was associated with a higher risk than non-A blood groups.33,34 The biologic mechanisms undergirding these findings may have to do with the ABO group per se (e.g., with the development of neutralizing antibodies against protein-linked N-glycans)36 or with other biologic effects of the identified variant,37-39 including the stabilization of von Willebrand factor.40,41 The ABOlocus holds considerable risk for population stratification,42 which is increased by the inclusion of randomly selected blood donors in the current study (for which there is an inherent risk of blood group O enrichment). Alignment of the allele frequencies at the ABO locus in our control population with those in several non–blood-donor control populations would suggest that this is not a major bias, and at least one study34 that tested for association with blood type used disease controls with no affiliation to blood donors.
The pragmatic aspects leading to the feasibility of this massive undertaking in a very short period of time during the extreme clinical circumstances of the pandemic imposed limitations that will be important to explore in follow-up studies. For example, to enable the recruitment of study participants, a bare minimum of clinical metadata was requested. For this reason, extensive genotype–phenotype elaboration of current findings could not be conducted, and adjustments for all potential sources of bias (e.g., underlying cardiovascular and metabolic factors relevant to Covid-19) could not be performed. Furthermore, we have limited information about the SARS-CoV-2 infection status in the control participants; this concern is mitigated by the fact that the presence of susceptible persons in the control group would only bias the tests toward the null. In addition, few restrictions were imposed during inclusion, which led to genotyped samples having to be excluded owing to differing ethnic groups (population outliers). Further exploration of current findings, both as to their usefulness in clinical risk profiling of patients with Covid-19 and toward a mechanistic understanding of the underlying pathophysiology, is warranted.

Supported by a philanthropic donation from Stein Erik Hagen and Canica; by a grant from the Deutsche Forschungsgemeinschaft Cluster of Excellence “Precision Medicine in Chronic Inflammation” (EXC2167); by a Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Covid-19 Biobank grant (to Dr. Valenti); by grants from the Italian Ministry of Health (RF-2016-02364358, to Dr. Valenti) and Ministero dell’Istruzione, dell’Università e della Ricerca project “Dipartimenti di Eccellenza 2018–2022” (D15D18000410001 to the Department of Medical Sciences, University of Turin; by a grant from the Spanish Ministry of Science and Innovation JdC fellowship (IJC2018-035131-I, to Dr. Acosta-Herrera); and by the GCAT Cession Research Project PI-2020-01. HLA typing was performed and supported by the Stefan-Morsch-Stiftung.
Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.
Dr. Ellinghaus and Ms. Degenhardt and Drs. Valenti, Franke, and Karlsen contributed equally to this article.
This article was published on June 17, 2020, at NEJM.org.
We thank all the patients who consented to participate in this study, and we express our condolences to the families of patients who died from Covid-19. We also thank the entire clinical staff during the outbreak situation at the different centers who were able to work on this scientific study in parallel with their clinical duties; all the members of the Humanitas Covid-19 Task Force for contributions to the recruitment of patients (see the Supplementary Notes section in Supplementary Appendix 1); Sören Brunak and Karina Banasik for discussions on the ABO association; Goncalo Abecasis and his team for providing the Michigan imputation server; Fabrizio Bossa and Francesca Tavano for contributions to control-sample acquisition; Maria Reig for help in the case-sample acquisition; the staff of the Basque Biobank in Spain for assistance in the acquisition of samples; the staff of GCAT|Genomes for Life, a cohort study of the Genomes of Catalonia, Institute for Health Science Research Germans Trias i Pujol, for data contribution; Alexander Eck, Jenspeter Horst, and Jens Scholz for supporting the HLA typing in the project; and the members of the ethics commissions, review boards, and consortia who fast-track reviewed our applications and enabled this rapid genetic discovery study.

Author Affiliations

From the Institute of Clinical Molecular Biology, Christian-Albrechts-University (D.E., F.D., J.K., S. May, M. Wendorff, L.W., F.U.-W., X.Y., A.T., A. Peschuck, C.G., G.H.-S., H.E.A., M.C.R., M.E.F.B., M. Schulzky, M. Wittig, N.B., S.J., T.W., W.A., M. D’Amato, A.F.), and University Hospital Schleswig-Holstein, Campus Kiel (N.B., A.F.), Kiel, the Institute for Cardiogenetics, University of Lübeck, Lübeck (J.E.), the German Research Center for Cardiovascular Research, partner site Hamburg–Lübeck–Kiel (J.E.), the University Heart Center Lübeck (J.E.), and the Institute of Transfusion Medicine, University Hospital Schleswig-Holstein (S.G.), Lübeck, Stefan-Morsch-Stiftung, Birkenfeld (M. Schaefer, W.P.), and the Research Group for Evolutionary Immunogenomics, Max Planck Institute for Evolutionary Biology, Plön (O.O., T.L.L.) — all in Germany; Novo Nordisk Foundation Center for Protein Research, Disease Systems Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen (D.E.); the Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute–Donostia University Hospital–University of the Basque Country (L.B., K.G.-E., L.I.-S., P.M.R., J.M.B.), Osakidetza Basque Health Service, Donostialdea Integrated Health Organization, Clinical Biochemistry Department (A.G.C., B.N.J.), and the Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute (M. D’Amato), San Sebastian, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III (L.B., M. Buti, A. Albillos, A. Palom, F.R.-F., B.M., L. Téllez, K.G.-E., L.I.-S., F.M., L.R., M.R.-B., M. Rodríguez-Gandía, P.M.R., M. Romero-Gómez, J.M.B.), the Departments of Gastroenterology (A. Albillos, B.M., L. Téllez, F.M., M. Rodríguez-Gandía), Intensive Care (R.P., A.B.O.), Respiratory Diseases (D.J., A.S., R.N.), Infectious Diseases (C.Q., E.N.), and Anesthesiology (D. Pestaña, N. Martínez), Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, University of Alcalá, and Histocompatibilidad y Biologia Molecular, Centro de Transfusion de Madrid (F.G.S.), Madrid, the Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus (M. Buti, A. Palom, L.R., M.R.-B.), Hospital Clinic, University of Barcelona, and the August Pi i Sunyer Biomedical Research Institute (J.F., F.A., E.S., J.F.-A., L.M., M.H.-T., P.C.), the European Foundation for the Study of Chronic Liver Failure (J.F.), Vall d’Hebron Institut de Recerca (A. Palom, F.R.-F., A.J., S. Marsal), and the Departments of Biochemistry (A.-E.G.-F., F.R.-F., A.C.-G., C.C., A.B.-G.), Intensive Care (R.F.), and Microbiology (T.P.), University Hospital Vall d’Hebron, the Immunohematology Department, Banc de Sang i Teixits, Autonomous University of Barcelona (E.M.-D.), Catalan Institute of Oncology, Bellvitge Biomedical Research Institute, Consortium for Biomedical Research in Epidemiology and Public Health and University of Barcelona, l’Hospitalet (V. Moreno), and Autonoma University of Barcelona (T.P.), Barcelona, Universitat Autònoma de Barcelona, Bellatera (M. Buti, F.R.-F., M.R.-B.), GenomesForLife–GCAT Lab Group, Germans Trias i Pujol Research Institute (A.C.N., I.G.-F., R.C.), and High Content Genomics and Bioinformatics Unit, Germans Trias i Pujol Research Institute (L. Sumoy), Badalona, Institute of Parasitology and Biomedicine Lopez-Neyra, Granada (J.M., M.A.-H.), the Digestive Diseases Unit, Virgen del Rocio University Hospital, Institute of Biomedicine of Seville, University of Seville, Seville (M. Romero-Gómez), and Ikerbasque, Basque Foundation for Science, Bilbao (M. D’Amato, J.M.B.) — all in Spain; the Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milan Bicocca (P.I., C.M.), Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico (D. Prati, G.B., A.Z., A. Bandera, A.G., A.L.F., A. Pesenti, C.P., F.C., F.M.-B., F.P., F.B., G.G., G. Costantino, L. Terranova, L. Santoro, L. Scudeller, M. Carrabba, M. Baldini, M.M., N. Montano, R.G., S.P., S. Aliberti, V. Monzani, S. Bosari, L.V.), the Department of Biomedical Sciences, Humanitas University (R.A., A. Protti, A. Aghemo, A. Lleo, E.M.P., G. Cardamone, M. Cecconi, V.R., S.D.), Humanitas Clinical and Research Center, IRCCS (R.A., A. Protti, A. Aghemo, A. Lleo, A.V., C.A., E.M.P., H.K., I.M., M. Cecconi, M. Ciccarelli, M. Bocciolone, P.P., P.O., P.T., S. Badalamenti, S.D.), University of Milan (A.Z., A. Bandera, A.G., A.L.F., A. Pesenti, F.M.-B., F.P., F.B., G.G., G. Costantino, M.M., N. Montano, R.G., S.P., S. Aliberti, S. Bosari, L.V.), and the Center of Bioinformatics, Biostatistics, and Bioimaging (M.G.V.) and the Phase 1 Research Center (M. Cazzaniga), School of Medicine and Surgery, and the Departments of Emergency, Anesthesia, and Intensive Care (G.F.), Pneumologia (P.F.), and Infectious Diseases (P.B.); University of Milano–Bicocca, Milan, the European Reference Network on Hepatological Diseases (P.I., C.M.) and the Infectious Diseases Unit (P.B.), San Gerardo Hospital, Monza, the Pediatric Departement and Centro Tettamanti–European Reference Network PaedCan, EuroBloodNet, MetabERN–University of Milano–Bicocca–Fondazione MBBM–Ospedale, San Gerardo (A. Biondi, L.R.B., M. D’Angiò), the Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (A. Latiano, O.P.), the Department of Medical Sciences, Università degli Studi di Torino, Turin (S. Aneli, G.M.), and the Italian Bone Marrow Donor Registry, E.O. Ospedali Galliera, Genoa (N.S.) — all in Italy; the Norwegian PSC Research Center, Department of Transplantation Medicine, Division of Surgery, Inflammatory Diseases, and Transplantation, and the Research Institute for Internal Medicine, Division of Surgery, Inflammatory Diseases, and Transplantation, Oslo University Hospital Rikshospitalet and University of Oslo (M.M.G., J.R.H., T.F., T.H.K.), and the Section for Gastroenterology, Department of Transplantation Medicine, Division for Cancer Medicine, Surgery, and Transplantation, Oslo University Hospital Rikshospitalet (J.R.H., T.F., T.H.K.), Oslo; the School of Biological Sciences, Monash University, Clayton, VIC, Australia (T.Z., M. D’Amato); Private University in the Principality of Liechtenstein (C.G.); the Institute of Biotechnology, Vilnius University, Vilnius, Lithuania (S.J.); and the Unit of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm (M. D’Amato).
Address reprint requests to Dr. Franke at the Institute of Clinical Molecular Biology and University Hospital of Schleswig-Holstein, Christian-Albrechts-University, Rosalind-Franklin-Str. 12, D-24105 Kiel, Germany, or at ; or to Dr. Karlsen at the Division of Surgery, Inflammatory Diseases, and Transplantation, Oslo University Hospital Rikshospitalet and University of Oslo, Postboks 4950 Nydalen, N-0424 Oslo, Norway, or at .
Dr. Franke serves as an author on behalf of the Covid-19 Host Genetics Initiative; members of the Initiative are listed in Supplementary Appendix 1, available at NEJM.org.

Supplementary Material

Supplementary Appendix 1PDF4831KB
Supplementary Appendix 2MS Excel16KB
Supplementary Appendix 3MS Excel10KB
Supplementary Appendix 4MS Excel14KB
Supplementary Appendix 5MS Excel12KB
Supplementary Appendix 6MS Excel13KB
Supplementary Appendix 7MS Excel15KB
Supplementary Appendix 8MS Excel40KB
Supplementary Appendix 9MS Excel92KB
Disclosure FormsPDF1920KB

LINK(S):
___

Contents of Ashley Biden’s Diary

National File Posts Contents of Ashley Biden’s Diary – Website Crashes By  Jim Hoft   Jan. 29, 2024 11:40 am  852 Comments Ashley Biden, und...