BACKGROUND

Evidence regarding the safety and efficacy of anticoagulant thromboprophylaxis among pediatric patients hospitalized for coronavirus disease 2019 (COVID-19) is limited. We sought to evaluate safety, dose-finding, and preliminary efficacy of twice-daily enoxaparin as primary thromboprophylaxis among children hospitalized for symptomatic COVID-19, including primary respiratory infection and multisystem inflammatory syndrome in children (MISC).

METHODS

We performed a phase 2, multicenter, prospective, open-label, single-arm clinical trial of twice-daily enoxaparin (initial dose: 0.5mg/kg per dose; max: 60mg; target anti-Xa activity: 0.20–0.49IU/mL) as primary thromboprophylaxis for children <18 years of age hospitalized for symptomatic COVID-19. Study endpoints included: cumulative incidence of International Society of Thrombosis and Haemostasis-defined clinically relevant bleeding; enoxaparin dose-requirements; and cumulative incidence of venous thromboembolism within 30-days of hospital discharge. Descriptive statistics summarized endpoint estimates that were further evaluated by participant age (±12 years) and clinical presentation.

RESULTS

Forty children were enrolled and 38 met analyses criteria. None experienced clinically relevant bleeding. Median (interquartile range) dose to achieve target anti-Xa levels was 0.5 mg/kg (0.48–0.54). Dose-requirement did not differ by age (0.5 [0.46–0.52] mg/kg for age ≥12 years versus 0.52 [0.49–0.55] mg/kg for age <12 years, P = .51) but was greater for participants with MISC (0.52 [0.5–0.61] mg/kg) as compared with primary COVID-19 (0.48 [0.39–0.51] mg/kg, P = .010). Two children (5.3%) developed central-venous catheter-related venous thromboembolism. No serious adverse events were related to trial intervention.

CONCLUSIONS

Among children hospitalized for COVID-19, thromboprophylaxis with twice-daily enoxaparin appears safe and warrants further investigation to assess efficacy.

What’s Known on This Subject:

Evidence is lacking regarding the safety and efficacy of anticoagulant thromboprophylaxis with twice-daily subcutaneous enoxaparin for pediatric patients hospitalized for symptomatic coronavirus disease 2019 (including primary respiratory infection and multisystem inflammatory syndrome in children).

What This Study Adds:

This open-label, multicenter, phase 2 clinical trial among children hospitalized for primary COVID-19 and multisystem inflammatory syndrome in children found twice-daily enoxaparin (0.5 mg/kg per dose; max of 60 mg per dose) to be safe, achieving target anti-Xa levels without observed clinically relevant bleeding or related-serious adverse events.

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has resulted in the infection of over 240 940 000 individuals and more than 4 903 900 deaths.1  Although initial reports suggested mild to moderate disease among hospitalized children,2  recent data from the United States indicate that over 26.5% require PICU admission, 15.4% noninvasive or invasive respiratory support, and 7.5% vasoactive infusion administration.3  Clinical presentations include primary acute respiratory infection (ie, coronavirus disease 2019 [COVID-19])4  and multisystem inflammatory syndrome in children (MIS-C).5,6  Both phenotypes are characterized by a dysregulated inflammatory response with marked coagulation activation, as evidenced by elevated plasma D-dimer levels, resulting in a prothrombotic state.712  The cumulative incidence of pediatric hospital-acquired venous thromboembolism (HA-VTE), including deep venous thrombosis (DVT) and/or pulmonary embolism, for hospitalized children with MIS-C are 6.5% and 2.1% for primary COVID-19.13  These estimates are substantively higher than those previously reported for hospitalized critically-ill (2%) and noncritically-ill (0.5%) children.1420 

Whereas the American College of Physicians recommends routine pharmacologic thromboprophylaxis for hospitalized adults without contraindications,21  the paucity of evidence from thromboprophylaxis clinical trials in pediatrics and validated risk models to predict HA-VTE limit the capacity to estimate a benefit from anticoagulant-based thromboprophylaxis weighed against the risk of thromboprophylaxis-related clinically-relevant bleeding.2224  As VTE represents a leading cause of preventable morbidity in hospitalized children, combinations of risk factors for heightened VTE risk2530  to inform the administration of mechanical and/or pharmacologic thromboprophylaxis are used, with the latter most commonly employing a low-dose regimen of twice-daily subcutaneous low molecular weight heparin (LMWH), enoxaparin.31,32  In the fall of 2020, the International Society on Thrombosis and Haemostasis (ISTH) recommended thromboprophylaxis for hospitalized children with COVID-19 (including MIS-C) who have markedly elevated D-dimer levels or superimposed clinical risk factors.33  Accompanying the recommendations were research priorities, including a call for rapid-activation of clinical trials assessing the safety and efficacy of pharmacologic thromboprophylaxis in children hospitalized for SARS-CoV-2 illness.

The primary aim of this phase 2, multicenter, prospective, open-label, single-arm clinical trial was to evaluate dose-finding, safety, and preliminary efficacy of twice-daily enoxaparin (starting dose, 0.5 mg/kg per dose, adjusted to achieve a target anti-Xa activity of 0.20–0.49 IU/mL) as the primary thromboprophylaxis in children younger than 18 years of age hospitalized for primary COVID-19 or MIS-C. The central hypothesis was that the cumulative incidence of ISTH-defined clinically relevant bleeding (primary safety endpoint) was ≤2.6% (upper limit of 90% confidence interval [90% CI], ≤11.8%) among children hospitalized for primary COVID-19 and MIS-C who received this thromboprophylaxis regimen.

The COVID-19 Anticoagulation in Children – Thromboprophylaxis (COVAC-TP) trial (www.clinicaltrials.gov, NCT04354155) was an investigator-initiated, multicenter, open-label, nonrandomized trial conducted at 8 children’s hospitals and academic medical centers in the United States and funded by the Johns Hopkins All Children’s Foundation from June 2020 through June 2021. The protocol (Supplemental Information 1) was approved by the institutional review board (IRB) at Johns Hopkins University (IRB 00247991) using a single IRB system for most centers, and independently reviewed at each participating site. Signed informed consent (and assent where appropriate) were required for all participants. The Clinical Coordinating Center and Data Coordinating Center were located at the Johns Hopkins All Children’s Institute for Clinical and Translational Research (St. Petersburg, FL). The trial monitoring plan is detailed in Supplemental Information 1. The trial was overseen by a Steering Committee and an Independent Data and Safety Monitoring Committee. All primary outcomes and adverse events were adjudicated by a Clinical Endpoint Adjudication Committee. The compositions of each study committee are provided in Supplemental Information 1.

Complete inclusion and exclusion criteria are provided in Supplemental Information 2. In summary, children <18 years of age (inclusive) hospitalized for symptomatic SARS-CoV-2-related illness including MIS-C and primary COVID-19 within an intensive care or acute care unit setting were eligible for enrollment. Main exclusion criteria were an expected length of stay (LOS) <2 days, treatment with therapeutic anticoagulation, concomitant aspirin at doses >5mg/kg per day, presence of clinically-relevant bleeding within 72 hours before enrollment, unwillingness to consent to participate, or any of the following laboratory-based exclusion criteria: platelet count <50 000/uL; prothrombin time ≥2 seconds above, or activated partial thromboplastin time ≥4 seconds above the upper limit of local age-dependent reference range; fibrinogen level <75 mg/dL; or estimated glomerular filtration rate <31 mL/min per 1.73 m2 as calculated using the Schwartz formula. Race and ethnicity (as self-identified by participants, using fixed categories) were recorded in accordance with National Institutes of Health requirements for tracking inclusion of minorities in clinical trials, and given racial and ethnic disparities in VTE incidence and outcomes, including hospitalized cases with COVID-19.13,34,35 

The protocol’s schedule of assessments is provided in Supplemental Table 4 and Supplemental Information 1. Patients received enoxaparin at a starting dose of 0.5 mg per kg (max starting dose: 60 mg per dose) subcutaneously every 12 hours, adjusted to achieve a 4 hour postdose anti-Xa level of 0.20 to 0.49 U/mL following the first or second dose based upon recommendations from the ISTH.33  For anti-Xa values <0.20 U/mL or ≥0.5 U/mL, enoxaparin dose was increased or decreased by 10% to 20%, respectively, and a repeat 4-hour postdose anti-Xa level was obtained. For patients who were started on enoxaparin twice-daily thromboprophylaxis before enrollment, based on the treating physician’s clinical judgement, the protocol did not require a change in dosing on-study if the anti-Xa level was in the protocol-specified target range. Serious adverse events, safety laboratory values, and pertinent clinical data were captured from patient enrollment through trial completion at 30 days (±2 days) postenrollment. Nonserious adverse events were not collected for this trial, for which an Investigational New Drug Application Waiver was granted by the US Food and Drug Administration.

The primary endpoint was safety—specifically, ISTH-defined clinically-relevant bleeding (ie, major plus clinically-relevant nonmajor bleeding)36  within 30-days of enrollment. Major bleeding included: (1) fatal bleeding; (2) clinically overt bleeding associated with a decrease in hemoglobin (Hgb) of at least 2 g/dL in a 24 hour period; (3) bleeding that is retroperitoneal, pulmonary, intracranial, or otherwise involves the central nervous system; and (5) bleeding that requires surgical intervention in an operating suite. Clinically-relevant nonmajor bleeding included: (1) overt bleeding for which a blood product is administered and that is not directly attributable to the patient’s underlying medical condition; and (2) bleeding that requires medical or surgical intervention to restore hemostasis, other than in an operating suite.

The secondary endpoint was the median twice-daily enoxaparin dose (mg/kg) required to achieve a 4 hour postdose antifactor Xa level of 0.20 to 0.49 anti-Xa U/mL. The cumulative incidence of HA-VTE within 30 days was defined as an exploratory efficacy endpoint. Clinically-relevant bleeding and recurrent VTE events reported by investigators were centrally adjudicated via a Clinical Endpoint Adjudication Committee.

The statistical analysis plan (Supplemental Information 1) was developed by the lead trial biostatistician (E.A.). Patient demographics and baseline characteristics were summarized with counts and percentages for categorical variables, and with means, standard deviations, medians, and interquartile ranges (IQR) as appropriate, for numeric variables. The primary analysis population for the study endpoints was prespecified as all patients received at least 2 doses of enoxaparin postenrollment and before hospital discharge. Cumulative incidences were calculated with corresponding exact (Clopper-Pearson) 90% confidence intervals (90% CIs). According to the protocol, we targeted an enrollment of 38 patients. We calculated with this sample size, the precision of estimates for various cumulative incidences of clinically-relevant bleeding (with corresponding 90% CIs) was as follows: 0 of 38, 0% (0% to 7.6%); 1 of 38, 2.6% (0% to 11.8%); and 2 of 38, 5.3% (1% to 15.7%). The secondary endpoint (enoxaparin dose requirement) was summarized by COVID-19 presentations (primary COVID-19 or MIS-C) with medians and corresponding IQRs. Medians were compared between age groups (<12 versus ≥12 years) and COVID-19 presentations (primary COVID-19 versus MIS-C) using the Kruskal-Wallis test. An age of 12 years was selected for subgroup analysis based on epidemiologic data demonstrating a peak in VTE incidence among adolescents.15,16,18  Two-sided P values of <.05 were considered to indicate statistical significance. Missing data were not imputed. Adverse events were descriptively analyzed in the safety population, defined as all patients who received at least 1 dose of enoxaparin postenrollment. Analyses were performed with SAS software, version 9.4 (SAS Institute, Cary, NC, USA).

A total of 40 patients from 8 participating United States sites were enrolled in the study, of which 38 were retained in the primary analysis population (Fig 1). One patient was withdrawn by parental request and another discharged from the hospital before receiving 2 doses of enoxaparin postenrollment. Clinical presentation was primary COVID-19 in 18 children and MIS-C in 20. The median age was 12.1 (range 1.3–17.5) years, 52.6% were male, and 47.4% were female. Table 1 summarizes demographic data, body mass index (BMI), and other baseline clinical characteristics by presentation; Table 2 provides these data by patient age <12 or ≥12 years (n = 19 each). As comparedwith children presenting with primary COVID-19, those with MIS-C were younger (median age [IQR]: 9.9 [7.3–12.6] versus 15.4 [12.5–17] years, P = .002), had lower BMI (median [IQR]: 20.1 [16.3–23.3] versus 32.1 [23.1–43.4] kg/m2, P < .001), and had higher D-dimer levels (median [IQR]: 3.4 [2.1–4.7] versus 0.8 [0.6–2] µg/mL, P < .001). Adolescents, when compared with younger children, had higher BMIs (median [IQR]: 31.3 [21.7–43.4] versus 18.8 [16.3–24.7] kg/m2, P < .001) and lower D-dimer levels (median [IQR] 1.2 [0.7–2] versus 3.4 [2.2–4.3] µg/mL, P = .001). Details on treatments employed for underlying COVID-19 disease are provided in Table 3.

FIGURE 1

Consolidated Standards of Reporting Trials flowchart for this single arm, open-label, phase 2 clinical trial with primary enoxaparin thromboprophylaxis in patients hospitalized for SARS-CoV-2.

FIGURE 1

Consolidated Standards of Reporting Trials flowchart for this single arm, open-label, phase 2 clinical trial with primary enoxaparin thromboprophylaxis in patients hospitalized for SARS-CoV-2.

Close modal
TABLE 1

Demographics, Anthropometrics, and Laboratory Data for Cohorts at Time of Study Enrollment for the Entire Study Sample and by COVID-19 Clinical Presentation (Primary Respiratory COVID-19 Infection Versus Multisystem Inflammatory Syndrome in Children)

CharacteristicPrimary COVID-19MIS-CP
(n = 18)(n = 20)
Age in years, median (IQR) 15.4 (12.5–17) 9.9 (7.3–12.6) .002 
Gender   .11 
 Male 7 (38.9) 13 (65)  
 Female 11 (61.1) 7 (35)  
Ethnicity   .38 
 Not Hispanic 10 (55.6) 15 (75)  
 Hispanic 7 (38.9) 5 (25)  
 Unknown or not reported 1 (5.6) 0 (0)  
Race   .31 
 Asian 0 (0) 1 (5)  
 Black or African American 5 (27.8) 7 (35)  
 More than 1 race 1 (5.6) 0 (0)  
 Native Hawaiian or Other Pacific Islander 1 (5.6) 0 (0)  
 Unknown 5 (27.8) 2 (10)  
 White or Caucasian 6 (33.3) 10 (50)  
Wt in kilograms, median (IQR) 86.8 (50.3–121.8) 37.2 (26.5–52.7) <.001 
BMI, median (IQR) 32.1 (23.1–43.4) 20.1 (16.3–23.3) <.001 
Noninvasive ventilation or supplemental oxygen at time of enrollment 14 (77.8) 12 (60%) .24 
Complete blood count data, median (IQR)    
 White blood cell count, K/µL 5.9 (3.6–10.3) 9.2 (5.2–13.8) .08 
 Hemoglobin, g/dL 12.8 (11.9–13.4) 9.8 (9.1–11.2) <.001 
 Platelet count, K/µL 181 (125–253) 145 (109–176) .54 
Coagulation data at screening, median (IQR)    
 Prothrombin time, sec 13.4 (12.6–14) 14.4 (13–16.2) .83 
 Partial thromboplastin time, sec 32.1 (30–34) 39 (33.1–43.5) .002 
 International normalized ratio 1 (0.9–1.2) 1.2 (1.1–1.3) .06 
 Fibrinogen, mg/dL 487 (388–525) 530 (367–580) .90 
 D-dimer, µg/mL 0.8 (0.6–2) 3.4 (2.1–4.7) <.001 
 Creatinine, mg/dL 0.6 (0.5–0.7) 0.6 (0.5–0.7) .41 
Presence of a central venous catheter    
 At study screening 5 (27.8) 5 (25) >.99 
 Day 1 postenrollment 5 (27.8) 9 (45) .27 
CharacteristicPrimary COVID-19MIS-CP
(n = 18)(n = 20)
Age in years, median (IQR) 15.4 (12.5–17) 9.9 (7.3–12.6) .002 
Gender   .11 
 Male 7 (38.9) 13 (65)  
 Female 11 (61.1) 7 (35)  
Ethnicity   .38 
 Not Hispanic 10 (55.6) 15 (75)  
 Hispanic 7 (38.9) 5 (25)  
 Unknown or not reported 1 (5.6) 0 (0)  
Race   .31 
 Asian 0 (0) 1 (5)  
 Black or African American 5 (27.8) 7 (35)  
 More than 1 race 1 (5.6) 0 (0)  
 Native Hawaiian or Other Pacific Islander 1 (5.6) 0 (0)  
 Unknown 5 (27.8) 2 (10)  
 White or Caucasian 6 (33.3) 10 (50)  
Wt in kilograms, median (IQR) 86.8 (50.3–121.8) 37.2 (26.5–52.7) <.001 
BMI, median (IQR) 32.1 (23.1–43.4) 20.1 (16.3–23.3) <.001 
Noninvasive ventilation or supplemental oxygen at time of enrollment 14 (77.8) 12 (60%) .24 
Complete blood count data, median (IQR)    
 White blood cell count, K/µL 5.9 (3.6–10.3) 9.2 (5.2–13.8) .08 
 Hemoglobin, g/dL 12.8 (11.9–13.4) 9.8 (9.1–11.2) <.001 
 Platelet count, K/µL 181 (125–253) 145 (109–176) .54 
Coagulation data at screening, median (IQR)    
 Prothrombin time, sec 13.4 (12.6–14) 14.4 (13–16.2) .83 
 Partial thromboplastin time, sec 32.1 (30–34) 39 (33.1–43.5) .002 
 International normalized ratio 1 (0.9–1.2) 1.2 (1.1–1.3) .06 
 Fibrinogen, mg/dL 487 (388–525) 530 (367–580) .90 
 D-dimer, µg/mL 0.8 (0.6–2) 3.4 (2.1–4.7) <.001 
 Creatinine, mg/dL 0.6 (0.5–0.7) 0.6 (0.5–0.7) .41 
Presence of a central venous catheter    
 At study screening 5 (27.8) 5 (25) >.99 
 Day 1 postenrollment 5 (27.8) 9 (45) .27 

Data are presented as n (%) unless otherwise indicated.

TABLE 2

Demographics, Anthropometrics, and Laboratory Data at the Time of Study Enrollment by Cohorts Defined by Participant Age (<12 y and ≥12 y of Age)

Characteristic≥12 y<12 yP
(n = 19)(n = 19)
Age in years, median (IQR) 15.4 (13.6–17.1) 8.4 (5.9–10.3) <.001 
Gender   .05 
 Male 7 (36.9) 13 (68.4)  
 Female 12 (63.1) 6 (31.6)  
Ethnicity   .50 
 Not Hispanic 11 (57.9) 14 (73.7)  
 Hispanic 7 (36.8) 5 (26.3)  
 Unknown or not reported 1 (5.3) 0 (0)  
Race   .51 
 Asian 0 (0) 1 (5.3)  
 Black or African American 5 (26.3) 7 (36.9)  
 More than 1 race 0 (0) 1 (5.3)  
 Native Hawaiian or other Pacific Islander 1 (5.3) 0 (0)  
 Unknown 5 (26.3) 2 (10.5)  
 White or Caucasian 8 (42.1) 8 (42.1)  
Wt in kilograms, median (IQR) 83.6 (56.2–121.8) 32.2 (26–49) <.001 
BMI, median (IQR) 31.3 (21.7–43.4) 18.8 (16.3–24.7) .004 
Noninvasive ventilation or supplemental oxygen at time of enrollment 12 (63.2) 14 (73.7) .49 
Complete blood count data, median (IQR)    
 White blood cell count, K/µL 6.3 (5.1–10.4) 8.4 (5.1–14.1) .16 
 Hemoglobin, g/dL 12.3 (11.2–13.2) 9.9 (8.9–11.9) .002 
 Platelet count, K/µL 148 (125–253) 158 (103–193) .78 
Coagulation data at screening, median (IQR)    
 Prothrombin time, sec 13.5 (12.6–15.3) 14.2 (13–15.2) .49 
 Partial thromboplastin time, sec 32 (29.4–35.8) 35.6 (33.1–41.4) .03 
 International normalized ratio 1.1 (1–1.3) 1.1 (1.1–1.2) .65 
 Fibrinogen, mg/dL 511 (374–587) 487 (370–554) .70 
 D-dimer, µg/mL 1.2 (0.7–2) 3.4 (2.2–4.3) .001 
 Creatinine, mg/dL 0.6 (0.5–0.7) 0.5 (0.4–0.7) .41 
Presence of a central venous catheter    
 At study screening 5 (26.3) 5 (26.3) >.99 
 Day 1 postenrollment 5 (26.3) 9 (47.4) .18 
Characteristic≥12 y<12 yP
(n = 19)(n = 19)
Age in years, median (IQR) 15.4 (13.6–17.1) 8.4 (5.9–10.3) <.001 
Gender   .05 
 Male 7 (36.9) 13 (68.4)  
 Female 12 (63.1) 6 (31.6)  
Ethnicity   .50 
 Not Hispanic 11 (57.9) 14 (73.7)  
 Hispanic 7 (36.8) 5 (26.3)  
 Unknown or not reported 1 (5.3) 0 (0)  
Race   .51 
 Asian 0 (0) 1 (5.3)  
 Black or African American 5 (26.3) 7 (36.9)  
 More than 1 race 0 (0) 1 (5.3)  
 Native Hawaiian or other Pacific Islander 1 (5.3) 0 (0)  
 Unknown 5 (26.3) 2 (10.5)  
 White or Caucasian 8 (42.1) 8 (42.1)  
Wt in kilograms, median (IQR) 83.6 (56.2–121.8) 32.2 (26–49) <.001 
BMI, median (IQR) 31.3 (21.7–43.4) 18.8 (16.3–24.7) .004 
Noninvasive ventilation or supplemental oxygen at time of enrollment 12 (63.2) 14 (73.7) .49 
Complete blood count data, median (IQR)    
 White blood cell count, K/µL 6.3 (5.1–10.4) 8.4 (5.1–14.1) .16 
 Hemoglobin, g/dL 12.3 (11.2–13.2) 9.9 (8.9–11.9) .002 
 Platelet count, K/µL 148 (125–253) 158 (103–193) .78 
Coagulation data at screening, median (IQR)    
 Prothrombin time, sec 13.5 (12.6–15.3) 14.2 (13–15.2) .49 
 Partial thromboplastin time, sec 32 (29.4–35.8) 35.6 (33.1–41.4) .03 
 International normalized ratio 1.1 (1–1.3) 1.1 (1.1–1.2) .65 
 Fibrinogen, mg/dL 511 (374–587) 487 (370–554) .70 
 D-dimer, µg/mL 1.2 (0.7–2) 3.4 (2.2–4.3) .001 
 Creatinine, mg/dL 0.6 (0.5–0.7) 0.5 (0.4–0.7) .41 
Presence of a central venous catheter    
 At study screening 5 (26.3) 5 (26.3) >.99 
 Day 1 postenrollment 5 (26.3) 9 (47.4) .18 

Data are presented as n (%) unless otherwise indicated.

TABLE 3

Concurrently Applied COVID-19 Therapies, Invasive Mechanical Ventilation Rates, and Hospital Length of Stay for Participants Enrolled Into Study Displayed by Cohorts Defined by Patient Age (±12 y) and Clinical Presentation (Primary COVID-19 Versus Multisystem Inflammatory Syndrome in Children).

Variables≥12 y<12 yPPrimary COVID-19MIS-CP
(n = 19)(n = 19)(n = 18)(n = 20)
Mechanical ventilation 4 (21.1) 2 (10.5) 0.66 5 (27.8) 1 (5) .08 
 Duration in days, median (IQR) 17 (8.59–21) 5.5 (2–9) 0.243 14 (9–20) 2 (2–2) .25 
Length of stay in days, median (IQR) 6 (3–7) 7 (4–11) 0.955 7 (3–13.5) 5.5 (4–9) .16 
SARS-CoV-2 treatment       
 Interleukin-6 antagonists 1 (5.3) 0 (0) >0.999 1 (5.6) 0 (0) .47 
 Remdesivir 8 (42.1) 4 (21.1) 0.163 12 (66.7 0 (0) <.001 
 Azithromycin 2 (10.5) 0 (0) 0.486 2 (11.1) 0 (0) .22 
 Interleukin-1 inhibitors 0 (0) 2 (10.5) 0.486 0 (0) 2 (10) .49 
 Nonsteroidal anti-inflammatory drugs 9 (47.4) 10 (52.6) 0.746 6 (33.3) 13 (65) .05 
 Intravenous immunoglobulin 7 (36.8) 13 (68.4) 0.051 2 (11.1) 18 (90) <.001 
 Systemic corticosteroids 14 (73.7) 17 (89.5) 0.405 12 (66.7) 19 (95) .04 
Variables≥12 y<12 yPPrimary COVID-19MIS-CP
(n = 19)(n = 19)(n = 18)(n = 20)
Mechanical ventilation 4 (21.1) 2 (10.5) 0.66 5 (27.8) 1 (5) .08 
 Duration in days, median (IQR) 17 (8.59–21) 5.5 (2–9) 0.243 14 (9–20) 2 (2–2) .25 
Length of stay in days, median (IQR) 6 (3–7) 7 (4–11) 0.955 7 (3–13.5) 5.5 (4–9) .16 
SARS-CoV-2 treatment       
 Interleukin-6 antagonists 1 (5.3) 0 (0) >0.999 1 (5.6) 0 (0) .47 
 Remdesivir 8 (42.1) 4 (21.1) 0.163 12 (66.7 0 (0) <.001 
 Azithromycin 2 (10.5) 0 (0) 0.486 2 (11.1) 0 (0) .22 
 Interleukin-1 inhibitors 0 (0) 2 (10.5) 0.486 0 (0) 2 (10) .49 
 Nonsteroidal anti-inflammatory drugs 9 (47.4) 10 (52.6) 0.746 6 (33.3) 13 (65) .05 
 Intravenous immunoglobulin 7 (36.8) 13 (68.4) 0.051 2 (11.1) 18 (90) <.001 
 Systemic corticosteroids 14 (73.7) 17 (89.5) 0.405 12 (66.7) 19 (95) .04 

Data are presented as n (%) unless otherwise indicated.

No primary safety endpoint (ie, clinically-relevant bleeding) events were observed (cumulative incidence, 0% [90% CI, 0% to 7.6%]). Enoxaparin dose requirement did not differ by age ≥12 vs <12 years (median [IQR]: 0.5 [0.46–0.52] versus 0.52 [0.49–0.55] mg/kg, P = .51) but was higher among those presenting with MIS-C compared with primary respiratory COVID-19 (0.52 [0.5–0.61] versus 0.48 [0.39–0.51] mg/kg, P = .010). Targeted prophylactic anti-Xa levels were achieved in 35 of 38 children (92.1%). Twenty-eight participants (74.7%) required no dosing adjustments to achieve target anti-Xa levels. Ten participants (26.3%) underwent a single dosing adjustment after initial anti-Xa levels, of which 8 (75%) represented dose increases per protocol.

Two participants with primary COVID-19 developed VTE (on days 1 and 7 postenrollment), yielding a cumulative incidence of 5.3% (90% CI, 1.0% to 15.7%). Both VTEs were central venous catheter-related DVTs of the upper extremities that occurred while receiving the enoxaparin regimen at the targeted prophylactic anti-Xa levels. The indications for central venous catheterization for both subjects were hemodynamic monitoring, frequent blood draws, and access to administer medications following invasive ventilation. No VTE were recorded after hospital discharge at time of 30-day follow up data collection. Of the 36 participants discharged from the hospital by the conclusion of the study period, 13 (36.1%) were prescribed outpatient thromboprophylaxis (aspirin, n = 11; rivaroxaban, n = 2). Participants prescribed aspirin at discharge all had MIS-C.

One death occurred during study in a participant with primary COVID-19. This was deemed by the principal investigator to be related to underlying COVID-19 illness and unrelated to study intervention; the independent data and safety monitoring committee concurred with this assessment. No other serious adverse events were observed.

This multicenter phase 2 trial assessed safety, dose-finding, and exploratory efficacy of subcutaneous enoxaparin twice-daily dosing at 0.5 mg/kg for primary thromboprophylaxis among children hospitalized for SARS-CoV-2-related illness. This trial represents 1 of few prospective pediatric investigations of thromboprophylaxis reported for hospitalized children, and the first among children hospitalized for COVID-19. These analyses demonstrated the safety of enoxaparin based on the absence of clinically-relevant bleeding events within the prespecified range of clinically-relevant bleeding events. Goal anti-Xa levels were achieved in 92.1% of participants with few dosing adjustments. Dosing did not appear to vary by age groups ≥12 versus <12 years of age. Children hospitalized for MIS-C required a statistically (but not clinically-relevant) greater dose to achieve goal anti-Xa levels than those with primary COVID-19. Participants with MIS-C had significantly elevated D-dimer levels (median of 3.4 µg/mL, IQR: 2.1–4.7) as compared with primary COVID-19 and the observed increased enoxaparin dose requirement may be related to the potent proinflammatory hypercoagulable state seen in MIS-C.5,6,13  Two children (5.3%) developed central venous catheter-related VTE during enoxaparin thromboprophylaxis. These findings question the intensity of enoxaparin exposure employed in this trial and preliminarily recommended by ISTH are sufficiently efficacious thromboprophylaxis regimens (versus therapeutic anticoagulation dosing) for VTE prevention in children hospitalized for COVID-19-related illness. Overall, for children hospitalized for COVID-19 related illness, primary thromboprophylaxis with twice-daily subcutaneous enoxaparin is safe with respect to the risk of clinically-relevant bleeding. Phase 3 trials of enoxaparin thromboprophylaxis are warranted in hospitalized children with primary COVID-19, MIS-C, and other proinflammatory conditions for which VTE risk is increased.

Severe COVID-19 infection results in a dysregulated inflammatory response, endothelial dysfunction, platelet activation, and a resultant prothrombotic state.412,37  This acquired coagulopathy is characterized by an elevation in thrombogenesis biomarkers (ie, D‐dimer and fibrinogen levels).38,39  Specific to MIS-C, routine as well as viscoelastic coagulation assays have revealed marked hypercoagulability and hyperfibrinolysis as compared with controls and those with COVID-19.40,41  Prothrombotic risk from MIS-C is enhanced by systemic inflammatory-acquired organ dysfunction, including cardiomyopathy, leading to changes in blood flow states.42,43  Similar to data reported in this manuscript, retrospective observational evidence suggests that the absence of thromboprophylaxis and presence of elevated inflammatory markers (ie, D-dimer levels) are independently associated with greater odds of HA-VTE in critically ill adults.9,4447  Additionally, patients with COVID-19 are at risk for clinically-relevant bleeding from consumptive coagulopathy, platelet dysfunction, and endotheliopathy.4851  Therefore, a balanced interpretation of clinical trial data assessing both thrombotic and bleeding risk is essential to distinguish optimal thromboprophylaxis strategies.

Recent randomized controlled trials among adults hospitalized with COVID-19 have evaluated the safety and efficacy of anticoagulation strategies, including therapeutic dosing, standard once-daily thromboprophylaxis (40 mg per day), and intermediate thromboprophylaxis (1 mg/kg per day).52  When considering the safety endpoint of clinically relevant bleeding, adult trial participants receiving thromboprophylaxis experienced comparable findings to this study ranging between 0.9% to 2.3%.5255  In contrast, trial participants exposed to therapeutic anticoagulation generally experienced greater rates of clinically relevant bleeding, ranging from 3.8% to 8%.5456 

While no clinically relevant bleeding events were observed in the present pediatric phase 2 multicenter trial of enoxaparin thromboprophylaxis, 2 participants with a central venous catheter developed VTE while receiving thromboprophylaxis. This rate is consistent with adult literature; overall, the incidence of major thromboembolism among adult trial participants receiving anticoagulants during hospitalization for COVID-19 ranged from 2% to 7.5%.5356  The rather broad range of efficacy findings may be explained by the intensity of anticoagulant regimens studied (ie, therapeutic versus various thromboprophylaxis strategies) and the distribution of acuity or complexity of patients studied. For example, in the HEP-COVID trial conducted by Spyropoulos and colleagues,53  adults hospitalized for COVID-19 with D-dimer levels >4 times the upper limit of normal were randomized to receive therapeutic versus prophylactic heparins. While the incidence of major thromboembolism and death was reduced (relative risk: 0.46, 95% CI:0.27–0.81) among non-ICU patients receiving therapeutic heparin, this was not true for adults hospitalized in an ICU setting (risk ratio: 0.92, 95% CI:0.62–1.39). Alternatively, an open-label, adaptive, randomized clinical trial among critically ill adults with COVID-19 showed reduced rates of pulmonary embolism for participants receiving therapeutic-dose heparinization versus standard-dose thromboprophylaxis (2.5% versus 7.5%).55  Taken together, the findings from prior trials in adults with COVID-19 and those from the present report support the need for phase 3 trials to definitively evaluate the efficacy and safety of the use and intensity of enoxaparin thromboprophylaxis in children hospitalized for COVID-19-related illness.

This study has several limitations. First, variations in underlying disease treatments among and within participating sites contributed to the heterogeneity of the study population, which limit the precision of the outcome estimates reported here. This reflects real-world experience that, throughout a rapidly-evolving SARS-CoV-2 pandemic, a variety of algorithms and agents were employed for the underlying treatment of primary COVID-19 and MIS-C. Second, as study enrollment began before knowledge of the MIS-C phenotype, this subgroup analysis was posthoc and should be interpreted with caution. Third, some children may have been hospitalized in the ICU setting as part of institutional cohorting and pandemic-related infection control measures, rather than a need for escalated care or monitoring. Fourth, although the majority of participants had an elevated D-dimer at enrollment, this was not an inclusion criterion as with previously cited adult trials. Additional prospective investigations are needed to elucidate potential relationships between prothrombotic markers (D-dimer, fibrinogen), clinical presentation (MIS-C versus COVID-19), and the development of HA-VTE. The presence of mechanical thromboprophylaxis (ie, sequential compression devices) in addition to primary anticoagulant thromboprophylaxis with enoxaparin was not assessed in this trial. The standard of care for all participating centers were to apply sequential compression devices to adolescents admitted to the ICU setting unless contraindicated. We cannot account for the potential impact of concurrent mechanical prophylaxis and future trials must consider comparative effectiveness. Although extended thromboprophylaxis regimens were prescribed, thromboprophylaxis following hospital discharge and its impact on subsequent VTE development as an outpatient was beyond the scope of this trial and represents a knowledge gap for further inquiry. Lastly, differences in dose requirements for children with MIS-C may have been influenced by factors related to participant BMI (ie, obesity) and pre-enrollment enoxaparin exposure.

In this open-label, multicenter, phase 2 clinical trial, we found twice-daily subcutaneous enoxaparin as the primary thromboprophylaxis among children hospitalized for symptomatic COVID-19-related illness to be safe without evidence of clinically relevant bleeding or other serious adverse events. Further investigation is warranted to definitively assess clinical efficacy.

We thank the COVAC-TP investigators (Supplemental Information 1); the clinical research coordinators and research nurses at all participating centers for their partnership in conducting the multicenter trial; the members of the Data and Safety Monitoring Committee; the teams at the Clinical Coordinating Center for Pediatric Multicenter Studies and Data Coordinating Center for Pediatric Multicenter Studies in the Johns Hopkins All Children’s Institute for Clinical and Translational Research (St. Petersburg, FL, USA); Jade Hanson, RN, CCRP for Trial Master maintenance and monitoring; Laurel McDevitt, MS, Deborah Martino, MA, and Fatima Tariq, MS, for project management; and all patients and parents who participated in the trial.

Dr Sochet conceptualized the study, designed the trial, provided overall trial oversight and coordination, contributed to data interpretation, and drafted the initial manuscript; Dr Morrison conceptualized the study, designed the trial, provided trial site oversight, and contributed to data interpretation; Drs Jaffray, Godiwala, Wilson, Thornburg, Bhat, Zia, Lawrence, Kudchadkar, and Russell assisted with study design, provided trial site oversight, and interpretation of results; Ms Hamblin provided overall trial oversight and coordinated data capture and quality assurance; Dr Streiff assisted with study design and interpretation of results; Dr Spyropoulos assisted with study design and interpretation of results; Dr Amankwah assisted with study design, provided data coordination and study oversight, performed statistical analyses, and assisted with interpretation of results; Dr Goldenberg conceptualized the study, designed the trial, provided overall trial oversight and coordination, contributed to data interpretation, and assisted with drafting of the initial manuscript; and all authors reviewed the manuscript before submission, approved the final manuscript as submitted and agree to be accountable for all aspects of the work.

FUNDING: This article is funded by the Johns Hopkins All Children’s Foundation Institutional Research Award.

CONFLICT OF INTEREST DISCLOSURES: The authors have indicated they have no conflicts of interest to disclose.

This trial is registered at www.ClinicalTrials.gov (NCT04354155).

The complete list of COVAC-TP Trial Investigators can be found in the Supplemental Information.

CI

confidence interval

DVT

deep vein thrombosis

HA-VTE

hospital acquired - venous thromboembolism

IQR

interquartile range

IRB

institutional review board

ISTH

International Society on Thrombosis and Haemostasis

LMWH

low molecular weight heparin

LOS

length of stay

MIS-C

multisystem inflammatory syndrome in children

1
The World Health Organization
.
WHO coronavirus (COVID-19) dashboard
.
Available at: https://covid19.who.int. Accessed October 19, 2021
2
Dong
Y
,
Mo
X
,
Hu
Y
, et al
.
Epidemiology of COVID-19 among children in China
.
Pediatrics
.
2020
;
145
(
6
):
e20200702
3
Delahoy
MJ
,
Ujamaa
D
,
Whitaker
M
, et al;
COVID-NET Surveillance Team
;
COVID-NET Surveillance Team
.
Hospitalizations associated with COVID-19 among children and adolescents – COVID – NET, 14 states, March 1, 2020-August 14, 2021
.
MMWR Morb Mortal Wkly Rep
.
2021
;
70
(
36
):
1255
1260
4
Borrelli
M
,
Corcione
A
,
Castellano
F
,
Fiori-Nastro
F
,
Santamaria
F
.
Coronavirus disease 2019 in children
.
Front Pediatr
.
2021
;
9
:
668484
5
Feldstein
LR
,
Rose
EB
,
Horwitz
SM
, et al;
Overcoming COVID-19 Investigators
;
CDC COVID-19 Response Team
.
Multisystem inflammatory syndrome in U.S. children and adolescents
.
N Engl J Med
.
2020
;
383
(
4
):
334
346
6
Son
MBF
,
Murray
N
,
Friedman
K
, et al;
Overcoming COVID-19 Investigators
.
Multisystem inflammatory syndrome in children – initial therapy and outcomes
.
N Engl J Med
.
2021
;
385
(
1
):
23
34
7
Cevik
M
,
Kuppalli
K
,
Kindrachuk
J
, %
Peiris
M
.
Virology, transmission, and pathogenesis of SARS-CoV-2
.
BMJ
.
2020
;
371
:
m3862
8
Wang
Y
,
Zhu
F
,
Wang
C
, et al
.
Children hospitalized with severe COVID-19 in Wuhan
.
Pediatr Infect Dis J
.
2020
;
39
(
7
):
e91
e94
9
Cohen
SL
,
Gianos
E
,
Barish
MA
, et al;
Northwell Health COVID-19 Research Consortium
.
Prevalence and predictors of venous thromboembolism or mortality in hospitalized COVID-19 patients
.
Thromb Haemost
.
2021
;
121
(
8
):
1043
1053
10
Giannis
D
,
Barish
MA
,
Goldin
M
, et al;
COVID-19 Consortium Group
.
Incidence of venous thromboembolism and mortality in patients with initial presentation of COVID-19
.
J Thromb Thrombolysis
.
2021
;
51
(
4
):
897
901
11
Liu
Y
,
Cai
J
,
Wang
C
,
Jin
J
,
Qu
L
.
A systematic review and meta-analysis of incidence, prognosis, and laboratory indicators of venous thromboembolism in hospitalized patients with coronavirus disease 2019
.
J Vasc Surg Venous Lymphat Disord
.
2021
;
9
(
5
):
1099
1111.e6
12
Hasan
SS
,
Radford
S
,
Kow
CS
,
Zaidi
STR
.
Venous thromboembolism in critically ill COVID-19 patients receiving prophylactic or therapeutic anticoagulation: a systematic review and meta-analysis
.
J Thromb Thrombolysis
.
2020
;
50
(
4
):
814
821
13
Whitworth
H
,
Sartain
SE
,
Kumar
R
, et al
.
Rate of thrombosis in children and adolescents hospitalized with COVID-19 or MIS-C
.
Blood
.
2021
;
138
(
2
):
190
198
14
Children’s Hospitals’ Association Solutions for Patient Safety
.
Our results: venous thromboembolism even rate
.
Available at: https://www.solutionsforpatientsafety.org/ourresults/. Accessed September 1, 2021
15
Sandoval
JA
,
Sheehan
MP
,
Stonerock
CE
,
Shafique
S
,
Rescorla
FJ
,
Dalsing
MC
.
Incidence, risk factors, and treatment patterns for deep venous thrombosis in hospitalized children: an increasing population at risk
.
J Vasc Surg
.
2008
;
47
(
4
):
837
843
16
Setty
BA
,
O’Brien
SH
,
Kerlin
BA
.
Pediatric venous thromboembolism in the United States: a tertiary care complication of chronic diseases
.
Pediatr Blood Cancer
.
2012
;
59
(
2
):
258
264
17
Tuckuviene
R
,
Christensen
AL
,
Helgestad
J
,
Johnsen
SP
,
Kristensen
SR
.
Pediatric venous and arterial noncerebral thromboembolism in Denmark: a nationwide population-based study
.
J Pediatr
.
2011
;
159
(
4
):
663
669
18
Raffini
L
,
Huang
YS
,
Witmer
C
,
Feudtner
C
.
Dramatic increase in venous thromboembolism in children’s hospitals in the United States from 2001 to 2007
.
Pediatrics
.
2009
;
124
(
4
):
1001
1008
19
Goldenberg
NA
.
Venous thromboembolism in children
.
Clin Adv Hematol Oncol
.
2019
;
17
(
6
):
326
329
20
Sochet
AA
,
Kiskaddon
A
,
Betensky
M
,
Goldenberg
N
.
Venous thromboembolism among critically ill children: a narrative review
.
Semin Thromb Hemost
.
2021
;
47
(
6
):
631
642
21
Qaseem
A
,
Chou
R
,
Humphrey
LL
, %
Starkey
M
,
Shekelle
P
;
Clinical Guidelines Committee of the American College of Physicians
.
Venous thromboembolism prophylaxis in hospitalized patients: a clinical practice guideline from the American College of Physicians
.
Ann Intern Med
.
2011
;
155
(
9
):
625
22
Faustino
EV
,
Hanson
S
,
Spinella
PC
, et al;
PROphylaxis against ThRombosis prACTice (PROTRACT) Study Investigators of the PALISI BloodNet
.
A multinational study of thromboprophylaxis practice in critically ill children
.
Crit Care Med
.
2014
;
42
(
5
):
1232
1240
23
Bigelow
AM
,
Flynn-O’Brien
KT
,
Simpson
PM
,
Dasgupta
M
,
Hanson
SJ
.
Multicenter review of current practices associated with venous thromboembolism prophylaxis in pediatric patients after trauma
.
Pediatr Crit Care Med
.
2018
;
19
(
9
):
e448
e454
24
Faustino
EV
,
Patel
S
,
Thiagarajan
RR
,
Cook
DJ
,
Northrup
V
,
Randolph
AG
.
Survey of pharmacologic thromboprophylaxis in critically ill children
.
Crit Care Med
.
2011
;
39
(
7
):
1773
1778
25
Arlikar
SJ
,
Atchison
CM
,
Amankwah
EK
, et al
.
Development of a new risk score for hospital-associated venous thromboembolism in critically-ill children not undergoing cardiothoracic surgery
.
Thromb Res
.
2015
;
136
(
4
):
717
722
26
Atchison
CM
,
Amankwah
E
,
Wilhelm
J
, et al
.
Risk factors for hospital-associated venous thromboembolism in critically ill children following cardiothoracic surgery or therapeutic cardiac catheterisation
.
Cardiol Young
.
2018
;
28
(
2
):
234
242
27
Amankwah
EK
,
Atchison
CM
,
Arlikar
S
, et al
.
Risk factors for hospital-sssociated venous thromboembolism in the neonatal intensive care unit
.
Thromb Res
.
2014
;
134
(
2
):
305
309
28
Yen
J
,
Van Arendonk
KJ
,
Streiff
MB
, et al
.
Risk factors for venous thromboembolism in pediatric trauma patients and validation of a novel scoring system: the risk of clots in kids with trauma score
.
Pediatr Crit Care Med
.
2016
;
17
(
5
):
391
399
29
Mahajerin
A
,
Jaffray
J
,
Branchford
B
, et al
.
Comparative validation study of risk assessment models for pediatric hospital-acquired venous thromboembolism
.
J Thromb Haemost
.
2020
;
18
(
3
):
633
641
30
Jaffray
J
,
Mahajerin
A
,
Branchford
B
, et al
.
A new risk assessment model for hospital-acquired venous thromboembolism in critically ill children: a report from the Children’s Hospital-Acquired Thrombosis Consortium
2021
;
23
(
1
):
e1
e9
31
Monagle
P
,
Chan
AKC
,
Goldenberg
NA
, et al
.
Antithrombotic therapy in neonates and children: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines
.
Chest
.
2012
;
141
(
2 Suppl
):
e737S
e801S
32
Monagle
P
,
Cuello
CA
,
Augustine
C
, et al
.
American Society of Hematology 2018 guidelines for management of venous thromboembolism: treatment of pediatric venous thromboembolism
.
Blood Adv
.
2018
;
2
(
22
):
3292
3316
33
Goldenberg
NA
,
Sochet
A
,
Albisetti
M
, et al;
Pediatric/Neonatal Hemostasis and Thrombosis Subcommittee of the ISTH SSC
.
Consensus-based clinical recommendations and research priorities for anticoagulant thromboprophylaxis in children hospitalized for COVID-19-related illness
.
J Thromb Haemost
.
2020
;
18
(
11
):
3099
3105
34
White
RH
,
Keenan
CR
.
Effects of race and ethnicity on the incidence of venous thromboembolism
.
Thromb Res
.
2009
;
123
(
Suppl 4
):
S11
S17
35
Aujesky
D
,
Long
JA
,
Fine
MJ
,
Ibrahim
SA
.
African American race was associated with an increased risk of complications following venous thromboembolism
.
J Clin Epidemiol
.
2007
;
60
(
4
):
410
416
36
Mitchell
LG
,
Goldenberg
NA
,
Male
C
, %
Kenet
G
,
Monagle
P
,
Nowak-Göttl
U
;
Perinatal and Paediatric Haemostasis Subcommittee of the SSC of the ISTH
.
Definition of clinical efficacy and safety outcomes for clinical trials in deep venous thrombosis and pulmonary embolism in children
.
J Thromb Haemost
.
2011
;
9
(
9
):
1856
1858
37
Goshua
G
,
Pine
AB
,
Meizlish
ML
, et al
.
Endotheliopathy in COVID-19-associated coagulopathy: evidence from a single-centre, cross-sectional study
.
Lancet Haematol
.
2020
;
7
(
8
):
e575
e582
38
Zaffanello
M
,
Piacentini
G
,
Nosetti
L
,
Ganzarolli
S
,
Franchini
M
.
Thrombotic risk in children with COVID-19 infection: a systematic review of the literature
.
Thromb Res
.
2021
;
205
:
92
98
39
Sharathkumar
AA
,
Faustino
EVS
, %
Takemoto
CM
.
How we approach thrombosis risk in children with COVID-19 infection and MIS-C
.
Pediatr Blood Cancer
.
2021
;
68
(
7
):
e29049
40
Ankola
AA
,
Bradford
VR
,
Newburger
JW
, et al
.
Coagulation profiles and viscoelastic testing in multisystem inflammatory syndrome in children
.
Pediatr Blood Cancer
.
2021
;
68
(
12
):
e29355
41
Al-Ghafry
M
,
Vagrecha
A
,
Malik
M
, et al
.
Multisystem inflammatory syndrome in children (MIS-C) and the prothrombotic state: coagulation profiles and rotational thromboelastometry in a MIS-C cohort
.
J Thromb Haemost
.
2021
;
19
(
7
):
1764
1770
42
Chang
JC
,
Matsubara
D
,
Morgan
RW
, et al
.
Skewed cytokine responses rather than the magnitude of the cytokine storm may drive cardiac dysfunction in multisystem inflammatory syndrome in children
.
J Am Heart Assoc
.
2021
;
10
(
16
):
e021428
43
Cherqaoui
B
,
Koné-Paut
I
,
Yager
H
, %
Bourgeois
FL
,
Piram
M
.
Delineating phenotypes of Kawasaki disease and SARS-CoV-2-related inflammatory multisystem syndrome: a French study and literature review
.
Rheumatology (Oxford)
.
2021
;
60
(
10
):
4530
4537
44
Santoro
F
,
Núñez-Gil
IJ
,
Viana-Llamas
MC
, et al
.
Anticoagulation therapy in patients with coronavirus disease 2019: results from a multicenter international prospective registry (health outcome predictive evaluation for corona virus disease 2019 [HOPE-COVID19])
.
Crit Care Med
.
2021
;
49
(
6
):
e624
e633
45
Mouhat
B
,
Besutti
M
,
Bouiller
K
, et al
.
Elevated D-dimers and lack of anticoagulation predict PE in severe COVID-19 patients
.
Eur Respir J
.
2020
;
56
(
4
):
2001811
46
Nopp
S
,
Moik
F
,
Jilma
B
,
Pabinger
I
, %
Ay
C
.
Risk of venous thromboembolism in patients with COVID-19: a systematic review and meta-analysis
.
Res Pract Thromb Haemost
.
2020
;
4
(
7
):
1178
1191
47
Nadkarni
GN
,
Lala
A
,
Bagiella
E
, et al
.
Anticoagulation, bleeding, mortality, and pathology in hospitalized patients with COVID-19
.
J Am Coll Cardiol
.
2020
;
76
(
16
):
1815
1826
48
Al-Samkari
H
,
Karp Leaf
RS
,
Dzik
WH
, et al
.
COVID-19 and coagulation: bleeding and thrombotic manifestations of SARS-CoV-2 infection
.
Blood
.
2020
;
136
(
4
):
489
500
49
Iba
T
,
Levy
JH
,
Connors
JM
,
Warkentin
TE
,
Thachil
J
,
Levi
M
.
The unique characteristics of COVID-19 coagulopathy
.
Crit Care
.
2020
;
24
(
1
):
360
50
Halaby
R
,
Cuker
A
,
Yui
J
, et al
.
Bleeding risk by intensity of anticoagulation in critically ill patients with COVID-19: a retrospective cohort study
.
J Thromb Haemost
.
2021
;
19
(
6
):
1533
1545
51
Demelo-Rodriguez
P
,
Farfán-Sedano
AI
,
Pedrajas
JM
, et al;
Riete-Bleeding Investigators
.
Bleeding risk in hospitalized patients with COVID-19 receiving intermediate- or therapeutic doses of thromboprophylaxis
.
J Thromb Haemost
.
2021
;
19
(
8
):
1981
1989
52
Talasaz
AH
,
Sadeghipour
P
,
Kakavand
H
, et al
.
Recent randomized trials of antithrombotic therapy for patients with COVID-19: JACC state-of-the-art review
.
J Am Coll Cardiol
.
2021
;
77
(
15
):
1903
1921
53
Spyropoulos
AC
,
Goldin
M
,
Giannis
D
, et al;
HEP-COVID Investigators
.
Efficacy and safety of therapeutic-dose heparin vs standard prophylactic or intermediate-dose heparins for thromboprophylaxis in high-risk hospitalized patients with COVID-19: the HEP-COVID randomized clinical trial
.
JAMA Intern Med
.
2021
;
181
(
12
):
1612
1620
54
Sadeghipour
P
,
Talasaz
AH
,
Rashidi
F
, et al;
INSPIRATION Investigators
.
Effect of intermediate-dose vs standard-dose prophylactic anticoagulation on thrombotic events, extracorporeal membrane oxygenation treatment, or mortality among patients With COVID-19 admitted to the intensive care unit: the INSPIRATION randomized clinical trial
.
JAMA
.
2021
;
325
(
16
):
1620
1630
55
Goligher
EC
,
Bradbury
CA
,
McVerry
BJ
, et al;
REMAP-CAP Investigators
;
ACTIV-4a Investigators
;
ATTACC Investigators
.
Therapeutic anticoagulation with heparin in critically ill patients with Covid-19
.
N Engl J Med
.
2021
;
385
(
9
):
777
789
56
Lopes
RD
,
de Barros E Silva
PGM
, %
Furtado
RHM
, et al;
ACTION Coalition COVID-19 Brazil IV Investigators
.
Therapeutic versus prophylactic anticoagulation for patients admitted to hospital with COVID-19 and elevated D-dimer concentration (ACTION): an open-label, multicentre, randomised, controlled trial
.
Lancet
.
2021
;
397
(
10291
):
2253
2263

Supplementary data