Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2020 Jun 11;34(8):2163–2172. doi: 10.1038/s41375-020-0910-1

Hematological features of persons with COVID-19

Qiubai Li 1,✉,#, Yulin Cao 1,#, Lei Chen 1,#, Di Wu 1,#, Jianming Yu 1,#, Hongxiang Wang 2,#, Wenjuan He 1,#, Li Chen 2,#, Fang Dong 3,#, Weiqun Chen 4,#, Wenlan Chen 1, Lei Li 5, Qijie Ran 6, Qiaomei Liu 7, Wenxiang Ren 1, Fei Gao 1, Zhichao Chen 1,, Robert Peter Gale 8, Yu Hu 1,
PMCID: PMC7289481  PMID: 32528042

Abstract

We studied admission and dynamic demographic, hematological and biochemical co-variates in 1449 hospitalized subjects with coronavirus infectious disease-2019 (COVID-19) in five hospitals in Wuhan, Hubei province, China. We identified two admission co-variates: age (Odds Ratio [OR] = 1.18, 95% Confidence Interval [CI] [1.02, 1.36]; P = 0.026) and baseline D-dimer (OR = 3.18 [1.48, 6.82]; P = 0.003) correlated with an increased risk of death in persons with COVID-19. We also found dynamic changes in four co-variates, Δ fibrinogen (OR = 6.45 [1.31, 31.69]; P = 0.022), Δ platelets (OR = 0.95 [0.90–0.99]; P = 0.029), Δ C-reactive protein (CRP) (OR = 1.09 [1.01, 1.18]; P = 0.037), and Δ lactate dehydrogenase (LDH) (OR = 1.03 [1.01, 1.06]; P = 0.007) correlated with an increased risk of death. The potential risk factors of old age, high baseline D-dimer, and dynamic co-variates of fibrinogen, platelets, CRP, and LDH could help clinicians to identify and treat subjects with poor prognosis.

Subject terms: Infectious diseases, Risk factors

Introduction

Most people with coronavirus infectious disease-2019 (COVID-19) have mild to moderate symptoms and recover after the appropriate medical intervention(s). However, 15–32 percent develop severe or critical COVID-19 with a case-fatality rate of 1–15% [16]. There are few data of hematological abnormalities in persons with COVID-19 [713]. We studied hematological co-variates in 1449 hospitalized persons with COVID-19 in five hospitals in Wuhan, China, interrogating correlations of admission parameters with COVID-19 outcomes.

Methods

Study design and subjects

We studied subjects in seven centers of five hospitals of Union Hospital (Central Hospital, Union West Hospital, and Union Tumor Hospital), Wuhan Central Hospital, General Hospital of Central Theater Command, PLA, Wuhan Third Hospital, and Wuhan Jin-Yin-Tan Hospital between January 20, and April 4, 2020. Subjects were studied on admission for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infection by quantitative real-time reverse transcriptase-polymerase chain reaction (qRT-PCR) of nasal and pharyngeal swabs and/or blood test for anti-SARS-CoV-2 IgG/IgM antibodies using a colloidal-gold-based 2019-nCoV IgG/IgM Detection Kit (Nanjing Vazyme Medical Technology, Nanjing, China). COVID-19 was diagnosed according to World Health Organization interim guidance and the Novel Coronavirus Pneumonia Diagnosis and Treatment Program of the National Health Commission of China [14, 15]. Severity of COVID-19 was graded as follows: (1) mild; mild clinical symptoms, no pneumonia on lung CT; (2) common: fever, cough and lung CT with pneumonia; (3) severe: respiratory distress (respiratory rate > 30 min−1, oxygen saturation (O2Sat) ≤ 93 percent at rest and/or ratio of arterial oxygen partial pressure to fractional inspired oxygen ≤300 mmHg (PaO2/FIO2); and (4) critical: aforementioned criteria of respiratory failure receiving mechanical ventilation, shock, and/or organ failure other than lung and/or intensive care unit (ICU) hospitalization [12, 15]. Subjects were treated as described below or according to Chinese government guidelines [1518]. The study was approved by the Ethics Committees of Union Hospital (2020-0095) and of Wuhan Central Hospital (2020-007). Written and oral informed consent from subjects was waived by the Ethics Committees.

Data collection

Electronic medical records of subjects including epidemiological, demographic, and laboratory data. Outcomes included: (1) death; (2) alive with two successive negative SARS-Cov-2 qRT-PCR tests; (3) interval from symptoms to hospital admission, discharge, or death; (4) negative qRT-PCT test; and (5) interval from admission to COVID-19 progression. Data collection forms were reviewed and verified independently by two researchers. A third researcher adjudicated discordances.

Statistical analysis

Categorical variables were described as frequency rates and percentages and continuous variables by median and interquartile range (IQR). Significance was tested by Kruskal–Wallis test or the Fisher exact test. A two-sided P < 0.05 was considered significant. Locally weighted regression and smoothing scatterplot was performed to equate a smooth curve about the relationship between time and values of laboratory parameters. Uni- and multivariable logistic regression models were used to interrogate co-variates associated with in-hospital death. Analyses were not been adjusted for multiple comparisons. Data were analyzed as of April 4, 2020.

Results

Baseline co-variates corelated with death from COVID-19

We analyzed data from 3559 consecutive subjects. One thousand seven hundred were excluded because of missing data of SARS-CoV-2 qRT-PCR or anti-SARS-CoV-2 IgG/IgM, 410 with co-morbidities and/or receiving drugs that would potentially affect bone marrow function (Supplementary Fig. 1). The resulting study cohort was 1449 subjects.

Seven hundred and thirty-three subjects (51%) were male. Median age was 57 years (IQR, 42–66 years), 576 (40%) were 60–79 years and 66 (5%), ≥80 years. Common signs and symptoms on admission included dry (485; 43%) or productive cough (551; 38%), fatigue (531; 37%), and shortness of breath (520; 37%). Eighty three subjects (6%) were smokers and 71 (6%), health care providers. Four subjects were exposed in the Hunan Wholesale Seafood Market and 94 (6%), close contact with persons with SARS-CoV-2-infection. Twenty nine subjects (2%) had mild COVID-19, 956 (66%), moderate, 347 (24%), severe and 117 (8%), critical. Lung computed tomography (CT) scan showed bilateral pneumonia in 1215 (87%), ground-glass opacity in 1041 (75%), patchy shadows in 579 (42%), and consolidation in 242 (18%). Eight hundred and twenty-six (57%) subjects had ≥1 abnormal liver function tests. One hundred and forty-four (10%), acute respiratory distress syndrome (ARDS) and 392 (27%), bacterial co-infections. Other complications had <10% frequencies including heart, acute kidney abnormalities, septic shock, and multiple organ failure. One hundred and fifty-six subjects (14%) received high-flow nasal cannula oxygen, 98 (7%), noninvasive ventilation (no intubation), 54 (4%), invasive ventilation (intubation), and four extracorporeal membrane oxygenation (ECMO). Median ICU stay was 12 days (IQR, 5–21 days). Median intervals from symptom onset to a negative SARS-CoV-2 qRT-PCR RNA, progression and death or discharge were 22 days (IQR 17–28 days), 10 days (IQR 7–14 days), and 30 days (IQR 23–37 days).

At the time of data lock 1327 (92%) subjects were alive and discharged; 122 (8%), died (Table 1). Subjects who died were significantly older than survivors (median 69 versus 55 years; P < 0.001) and more likely male (74% versus 48%; P < 0.001), with more frequent fatigue (54% versus 35%; P < 0.001), chills (26% versus 15%; P < 0.001), productive cough (50% versus 37%, P = 0.006), shortness of breath (66% versus 35%; P < 0.001), a higher temperature on admission (median 36.8 °C; IQR 36.5–37.8 °C versus median 36.6 °C; IQR 36.4–37.0 °C; P < 0.001), bilateral pneumonia (95% versus 86%; P = 0.01), and/or consolidation (35% versus 16%, P < 0.001) on lung CT scan but similar dry cough (44% versus 43%, P = 0.824), diarrhea (both 13%; P = 0.897), and myalgia (20% versus 17%; P = 0.502).

Table 1.

Baseline characteristics of survivors and non-survivors with COVID-19.

Total
n = 1449
Alive
n = 1327
Died
n = 122
P value
Characteristics
Age, median (IQR), years 57 (42, 66) 55 (41, 65) 69 (63, 78) <0.001
Age distribution
 <40 years 306 (21) 305 (23) 1 (0.8)
 40–59 years 501 (35) 481 (36) 20 (16)
 60–79 years 576 (40) 499 (38) 77 (63)
 ≥80 years 66 (5) 42 (3) 24 (20)
Female sex 716 (49) 684 (52) 32 (26) <0.001
Smoking history 83 (6) 66 (5) 17 (15) <0.001
Health care provider 71 (6) 69 (7) 2 (2) 0.038
Exposure history 0.026
 Huanan Seafood Market 4 (0.28) 2 (0.15) 2 (2)
 Close contact with patients 94 (6) 89 (7) 5 (4)
Signs and symptoms
 Temperature (°C) 36.7 (36.4, 37.1) 36.6 (36.4, 37.0) 36.8 (36.5, 37.8) <0.001
 Shortness of breath 520 (37) 456 (35) 64 (66) <0.001
 Dry cough 485 (43) 437 (43) 48 (44) 0.824
 Wet cough 551 (38) 491 (37) 60 (50) 0.006
 Fatigue 531 (37) 466 (35) 65 (54) <0.001
 Nausea or vomiting 88 (6) 83 (6) 5 (4) 0.43
 Diarrhea 186 (13) 170 (13) 16 (13) 0.897
 Chill 226 (16) 194 (15) 32 (26) <0.001
 Runny nose 30 (2) 28 (2) 2 (1.6) 0.999
 Myalgia 255 (18) 231 (17) 24 (20) 0.502
 Headache 89 (6) 84 (6) 5 (4) 0.43
Staging <0.001
 Mild 29 (2) 29 (2) 0 (0)
 Moderate 956 (66) 951 (72) 5 (4)
 Severe 347 (24) 332 (25) 15 (12)
 Critical 117 (8) 15 (1) 102 (84)
Imaging features
 Bilateral pneumonia 1215 (87) 1117 (86) 98 (95) 0.01
 Consolidation 242 (18) 207 (16) 35 (35) <0.001
 Ground-glass opacity 1041 (75) 974 (76) 67 (67) 0.059
 Patchy shadows 579 (42) 532 (41) 47 (47) 0.218
Complications
 ARDS 144 (10) 37 (3) 107 (88) <0.001
 Bacterial infections 392 (27) 288 (22) 104 (85) <0.001
 Septic shock 45 (4) 1 (0.1) 44 (40) <0.001
 Acute kidney injury 49 (3) 7 (0.5) 42 (34) <0.001
 Cardiac injury 125 (9) 58 (4) 67 (55) <0.001
 Liver damage 826 (57) 721 (54) 105 (86) <0.001
 Gastrointestinal bleeding 20 (1) 4 (0.3) 16 (13) <0.001
 Coagulopathy 28 (2) 0 (0) 28 (23) <0.001
 Multiple organ failure 72 (5) 3 (0.2) 69 (57) <0.001
Treatments
 Antibiotics 1203 (85) 1084 (83) 119 (98) <0.001
 Antimycotics 44 (3) 17 (1) 27 (22) <0.001
 Oseltamivir 604 (42) 564 (43) 40 (33) 0.037
 Umifenovir 1099 (76) 1004 (76) 95 (78) 0.688
 Lopinavir and Ritonavir 276 (24) 232 (23) 44 (40) <0.001
 Interferon 297 (21) 269 (20) 28 (23) 0.483
 Corticosteroids 576 (40) 471 (35) 105 (86) <0.001
 Intravenous immunoglobin 381 (28) 314 (25) 67 (55) <0.001
 High-flow nasal cannula oxygen therapy 156 (14) 59 (6) 97 (88) <0.001
 Noninvasive mechanical ventilation 98 (7) 21 (2) 77 (63) <0.001
 Invasive mechanical ventilation 54 (4) 7 (0.5) 47 (39) <0.001
 ECMO 4 (0.4) 1 (0.1) 3 (3) 0.003
Outcomes
 ICU admission 63 (4) 23 (2) 40 (33) <0.001
 Time from illness onset to ICU admission, median (IQR), days 14 (10, 19) 10 (8, 18) 15 (12, 20) 0.141
 ICU length of stay, median (IQR), days 12 (5, 21) 11 (8, 21) 12 (4, 21) 0.619
 Time from illness onset to repeated negatively tests of SARS-CoV-2, median (IQR), days 22 (17, 28) 22 (17, 28) 19 (12, 26) 0.066
 Time from illness onset to admission, median (IQR), days 10 (7, 15) 10 (7, 16) 10 (7, 12) 0.017
 Time from illness onset to progression, median (IQR), days 10 (7, 14) 10 (6, 14) 12 (9, 19) 0.001
 Time from illness onset to outcome, median (IQR), days 30 (23, 37) 30 (24, 38) 21 (15, 29) <0.001
 Time from diagnosis to outcome, median (IQR), days 19 (13, 27) 20 (13, 27) 11 (5, 17) <0.001
 Time from admission to outcome, median (IQR), days 18 (13, 23) 18 (13, 23) 11 (6, 20) <0.001

IQR interquartile ranges, ARDS acute respiratory distress syndrome, ECMO extracorporeal membrane oxygenation, ICU intensive care unit, SARS-CoV-2 severe acute respiratory syndrome coronavirus 2.

Median interval from symptom(s) onset to admission was 10 days in survivors (IQR 7–16 days) versus 10 days (IQR 7–12 days; P = 0.017) in non-survivors. Median interval from admission to progression in survivors was briefer (10 days [IQR 6–14 days] versus 12 days [IQR 9–19; P = 0.001). Median interval from onset to ICU admission was 14 days (IQR 10–19 days) and median ICU stay, 12 days (IQR 5–21 days) with no significate difference between survivors and non-survivors (P = 0.141 and P = 0.619). Subjects who died were more likely to have complications (122 [100%] versus 741 [56%]; P < 0.001) including acute respiratory distress syndrome (ARDS; 107 [88%] versus 37 [3%]; P < 0.001), liver function test abnormalities (105 [86%] versus 721 [54%]; P < 0.001) and bacterial infections (104 [85%] versus 288 [22%]; P < 0.001). More subjects dying received high-flow nasal cannula oxygen therapy (97 [88%] versus 59 [6%]; P < 0.001). Four subjects received ECMO including one survivor.

Blood hematological co-variates of survivors and non-survivors

We first compared admission hematological co-variates between survivors and non-survivors. Subjects who died had a higher median WBC (8 × 10E + 9/L [IQR 6–11 × 10E + 9/L] versus 5 × 10E + 9/L [IQR 4–7 × 10E + 9/L]; P < 0.001), higher median neutrophils (7 × 10E + 9/L [IQR 5–10 × 10E + 9/L] versus 3 × 10E + 9/L [IQR 2–4 ×10E + 9/L]; P < 0.001), lower median lymphocytes (0.5 × 10E + 9/L [IQR 0.4–0.8 × 10E + 9/L] versus 1.2 × 10E + 9/L [IQR 0.9–1.7 × 10E + 9/L]; P < 0.001) and lower median platelets (166 × 10E + 9/L [IQR 109–223 × 10E + 9/L] versus 208 × 10E + 9/L [IQR 164–268 × 10E + 9/L]; P < 0.001) compared with survivors (Table 2).

Table 2.

Blood hematological co-variates of survivors and non-survivors with COVID-19.

N Total Alive Died P value
Hemoglobin, g/L (115–150)
 Baseline 1437 129 (119–139) 128 (119–139) 131 (122–143) 0.068
 Max 1101 132 (122–142) 131 (126–146) 136 (122–142) 0.049
 Min 1101 119 (108–131) 120 (110–131) 105 (78–126) <0.001
White blood cell, ×10E + 9/L (3.5–10)
 Baseline 1420 5 (4–7) 5 (4–7) 8 (6–11) <0.001
 Max 1421 7 (5–9) 7 (5–8) 16 (12–21) <0.001
 Min 1421 5 (4–6) 5 (4–6) 6 (4–9) <0.001
Neutrophil, ×10E + 9/L (1.8–6.3)
 Baseline 1417 3 (2–5) 3 (2–4) 7 (5–10) <0.001
 Max 1421 5 (3–7) 4 (3–6) 14 (11–20) <0.001
 Min 1421 3 (2–4) 3 (2–4) 5 (3–8) <0.001
Lymphocyte, ×10E + 9/L (1.1–3.2)
 Baseline 1440 1.2 (0.8–1.6) 1.2 (0.9–1.7) 0.5 (0.4–0.8) <0.001
 Min 1411 1.0 (0.7–1.4) 1.1 (0.8–1.5) 0.3 (0.2–0.5) <0.001
Monocyte, ×10E + 9/L (0.1–0.6)
 Baseline 1408 0.4 (0.3–0.53) 0.4 (0.3–0.5) 0.3 (0.2–0.5) <0.001
 Max 1419 0.5 (0.4–0.7) 0.5 (0.4–0.7) 0.5 (0.4–0.8) 0.305
 Min 1419 0.3 (0.2–0.4) 0.3 (0.3–0.4) 0.2 (0.1–0.3) <0.001
Platelet, ×109 E + 9/L (125–350)
 Baseline 1415 206 (159–264) 208 (164–268) 166 (109–223) <0.001
 Max 1420 258 (204–325) 263 (208–331) 190 (134–255) <0.001
 Min 1420 176 (135–224) 180 (143–226) 80 (39–147) <0.001

Data are presented as medians (interquartile ranges, IQR). p values were calculated by Mann–Whitney U test, χ² test, or Fisher’s exact test, as appropriate.

Blood lymphocyte subsets

We next analyzed baseline blood lymphocyte subsets between subjects who died and survivors (Table 3). Subjects who died had lower median CD3-positive cells (140 × 10E + 9/L [IQR 75–190 × 10E + 9/L] versus 381 × 10E + 9/L [IQR 2–918 × 10E + 9/L]; P = 0.023), CD3-positive/CD4-positive cells (71 × 10E + 9/L [IQR 46–107 × 10E + 9/L] versus 227 × 10E + 9/L [IQR 1–487 × 10E + 9/L]; P = 0.036), CD3-positive/CD8-positive cells (49 × 10E + 9/L [IQR 14–64 × 10E + 9/L] versus 141 × 10E + 9/L [IQR, 1–308 × 10E + 9/L]; P = 0.023) and lower median proportions of CD3-positive cells (59% [IQR 50–67%] versus 72% [IQR 63–78%]; P < 0.001), median proportions of CD3-positive/CD8-positive cells (14% [IQR 10–18%] versus 24% [IQR 19–30%]; P < 0.001) and median proportions of B cells (16% [IQR 10–28%] versus 12% [IQR 9–17%]; P = 0.022). There were no significant differences in median proportions of CD3-positive/CD4-positive T- or natural-killer (NK)-cells, nor in concentrations of B cells or NK-cells between survivors and non-survivors.

Table 3.

The lymphocyte subsets of peripheral blood in of survivors and non-survivors with COVID-19.

Lymphocyte subset N Total Alive Died P value
CD3+ (%) 579 72 (62–78) 72 (63–78) 59 (50–67) <0.001
CD3 concentration × 10E + 9/L 246 359 (2–901) 381 (2–918) 140 (75–190) 0.023
CD3+CD4+ (%) 579 41 (33–48) 41 (33–48) 37 (28–48) 0.443
CD3+CD4+ concentration × 10E + 9/L 246 200 (1–481) 227 (1–487) 71 (46–107) 0.036
CD3+CD8+ (%) 579 23 (18–30) 24 (19–30) 14 (10–18) <0.001
CD3+CD8+ concentration × 10E + 9/L 246 119 (1–302) 141 (1–308) 49 (14–64) 0.023
NK cell (%) 415 10 (6–17) 10 (6–17) 10 (5–12) 0.29
NK cell concentration × 10E + 9/L 246 76 (0.4–185) 77 (0.4–188) 41 (19–102) 0.453
B lymphocyte (%) 415 13 (9–18) 12 (9–17) 16 (10–28) 0.022
B lymphocyte concentration × 10E + 9/L 246 96 (0.3–178) 98 (0.3–188) 55 (22–91) 0.136
CD4+/CD8+ ratio
Max 577 2 (1.4–2.6) 1.9 (1.4–2.6) 3.0 (1.9–4.5) <0.001
Min 489 1.6 (1.2–2) 1.57 (1.2–2) 1.6 (1.3–2.3) 0.336

Data are median (IQR), n (%), or n/N (%). Cell count at presentation (cells/ul). p values were calculated by Mann–Whitney U test, χ2 test, or Fisher’s exact test, as appropriate.

NK cell natural killer cell

Clotting co-variates

Baseline and maximum values of prothrombin time, activated partial thromboplastin time, and D-dimer concentrations were significantly higher in subjects who died compared with survivors. In contrast, fibrinogen concentration was higher at baseline in subject who died (median 4.3 g/L [IQR, 3.2–5.2 g/L] versus 3.6 g/L [IQR 2.9–4.5 g/L); P < 0.001) but had lower minimum values (2.6 g/L [IQR 1.7–3.9 g/L] versus 3.2 g/L [IQR 2.6–3.9 g/L]; P < 0.001; Table 4).

Table 4.

Clotting factor levels of survivors and non-survivors with COVID-19.

N Total Alive Died P value
PT, s (11–16)
 Baseline 1055 13 (12–13) 13 (12–13) 14 (13–15) <0.001
 Max 1035 13 (12–14) 13 (12–14) 17 (14–20) <0.001
APTT, s (28–43.5)
 Baseline 1055 34 (30–38) 34 (30–37) 35 (300–40) 0.019
 Max 1289 34 (30–38) 34 (30–37) 40 (34–50) <0.001
D-dimer, mg/L (<0.5)
 Baseline 1239 0.4 (0.2–0.9) 0.4 (0.2–0.8) 3.6 (0.9–8) <0.001
 Max 1262 0.6 (0.2–1.6) 0.5 (0.2–1) 8 (6–8) <0.001
Fibrinogen, g/L (2–4)
 Baseline 1304 3.7 (2.9–4.6) 3.6 (2.9–4.5) 4.3 (3.2–5.2) <0.001
 Min 976 3.2 (2.5–3.9) 3.2 (2.6–3.9) 2.6 (1.7–3.9) <0.001

PT prothrombin time, APTT activated partial thromboplastin time.

Inflammatory and biochemical co-variates

Median admission concentrations of C-reactive protein (CRP) (93 mg/L [IQR 58–125 mg/L] versus 9 mg/L [IQR 3–30 mg/L]; P < 0.001), procalcitonin (0.2 ng/ml [IQR 0.12–0.6 ng/ml] versus 0.05 ng/ml [IQR 0.04–0.1 ng/ml]; P < 0.001) and lactate dehydrogenase (LDH) (470 U/L [IQR 359–599 U/L] versus 199 U/L [IQR 161–258 U/L]; P < 0.001) were significantly higher in subjects who died compared with survivors (Table 5). Subjects who died were more likely to have abnormal heart, liver, and/or kidney function and to have higher admission median concentrations of interleukin-6 (IL-6; 71 pg/ml [IQR 29–442 pg/ml] versus 9 pg/ml [IQR 4–30 pg/ml]; P < 0.001) and interleukin-10 (IL-10; 11 pg/ml IQR [6–30 pg/ml] versus 4 pg/ml [IQR 3–5 pg/ml]; P < 0.001). There were no significant differences in concentrations of interleukins-2 (IL-2) or -4 (IL-4), tumor necrosis factor (TNF)-α, or interferon (IFN)-γ between survivors and non-survivors.

Table 5.

Biochemical parameters and inflammatory cytokines of survivors and non-survivors with COVID-19.

N Total Alive Died P value
CRP, mg/L (<8)
 Baseline 1046 11 (3, 44) 9 (3, 30) 93 (58, 125) <0.001
 Max 1063 14 (4, 55) 11 (3, 38) 140 (110, 181) <0.001
Procalcitonin, ng/ml (<0.5)
 Baseline 1273 0.05 (0.05, 0.1) 0.05 (0.04, 0.1) 0.2 (0.12, 0.6) <0.001
 Max 1065 0.07 (0.04, 0.1) 0.06 (0.04, 0.1) 1.2 (0.4, 4) <0.001
LDH, U/L (109–245)
 Baseline 1338 207 (165, 283) 199 (161, 258) 470 (359, 599) <0.001
 Max 1354 215 (174, 302) 207 (169, 271) 707 (509, 1154) <0.001
Ferritin, ng/ml (4.6–204) 231 542 (226, 1207) 446 (191, 906) 1584 (1196, 2000) <0.001
ALT, U/L (5–35) 1429 39 (22, 68) 37 (21, 66) 62 (34, 150) <0.001
AST, U/L (8–40) 1428 31 (22, 49) 30 (22, 44) 68 (45, 143) <0.001
Total bilirubin, μmol/L (5.1–19) 1245 14 (10, 19) 13 (10, 17) 25 (16, 39) <0.001
Creatine kinase, U/L (26–140) 1151 87 (54, 150) 81 (52, 130) 253 (104, 656) <0.001
BNP, pg/ml (<100) 627 50 (14, 160) 39 (12, 115) 454 (116, 1377) <0.001
Myoglobin, ng/ml (<140) 718 32 (21, 60) 29 (21, 49) 476 (147, 1200) <0.001
Troponin I, ng/L (<26.2) 830 3 (1, 10) 2 (0.9, 7) 212 (48, 1011) <0.001
BUN, mmol/L (2.9–8.2) 1414 5 (4, 6) 5 (4, 6) 14 (8, 23) <0.001
Scr, μmol/L (44–106) 1414 70 (59, 83) 69 (59, 81) 100 (71, 211) <0.001
IL-2, pg/ml (0.1–4.1) 344 3 (3, 4) 3 (3, 4) 3 (3, 5) 0.849
IL-4, pg/ml (0.1–3.2) 380 3 (2, 4) 3 (2, 4) 2 (2, 4) 0.804
IL-6, pg/ml (0.1–2.9) 659 10 (4, 37) 9 (4, 30) 71 (29, 442) <0.001
IL-10, pg/ml (0.1–5) 380 4 (3, 6) 4 (3, 5) 11 (6, 30) <0.001
TNF α, pg/ml (0.1–23) 380 3 (2, 6) 4 (2, 6) 3 (2, 4) 0.183
IFN γ, pg/ml (0.1–18) 380 3 (2, 4) 3 (2, 4) 3 (2, 4) 0.88

CRP C-reactive protein, LDH lactate dehydrogenase, ALT alanine aminotransferase, AST aspartate aminotransferase, BNP brain natriuretic peptide, BUN blood urea nitrogen, Scr serum creatinine, IL interleukin, TNF tumor necrosis factor, IFN interferon.

Dynamic changes in hematological co-variate

Next, we studied dynamic changes in hematological co-variates between survivors and non-survivors in 390 subjects from Wuhan Third Hospital with daily determinations (Supplementary Table 1). Subjects who died had higher concentrations of WBCs, neutrophils, D-dimmer, PT, LDH, and CRP but lower concentrations of lymphocytes and platelets throughout their hospitalization. These dynamic changes are displayed in Fig. 1.

Fig. 1. Dynamic changes of hematological variables in patients with COVID-19 during hospitalization.

Fig. 1

The Y-axis “value” include units of all above data: ×10E + 9/L for white bloods cells, neutrophils, lymphocytes, monocytes, and platelets; g/L for hemoglobin, fibrinogen (FIB); mg/L for D-dimer; s for prothrombin time (PT), activated partial thromboplastin time (APTT); U/L for lactate dehydrogenase (LDH); ng/ml for procalcitonin (PCT); mg/L for C-reactive protein (CRP). Data are presented as medians (interquartile ranges, IQR).

Uni- and multivariable analyses

We analyzed admission hematological co-variates and their multiple measurements considering changes (Δ = Max − Min, Max − baseline, or baseline − Min) correlated with risk of death in all subjects (Supplementary Table 2). In a multivariable logistic regression model, age (Odds Ratio [OR] = 1.18 [1.02, 1.36]; P = 0.026), baseline D-dimer (OR = 3.18 [1.48, 6.82]; P = 0.003), Δ fibrinogen (OR = 6.45 [1.31, 31.69]; P = 0.022), Δ platelets (OR = 0.95 [0.90–0.99]; P = 0.029), Δ CRP (OR = 1.09 [1.01, 1.18]; P = 0.037) and Δ LDH (OR = 1.03 [1.01, 1.06]; P = 0.007) correlated with an increased risk of death (Table 6).

Table 6.

Multivariate analysis of hematological co-variates associated with in-hospital death of patients with COVID-19.

Odds Ratio 95% Confidence Interval P value
Age (years) 1.18 (1.02–1.36) 0.026
Baseline D-dimer (mg/L) 3.18 (1.48–6.82) 0.003
Δ Platelet (×10E + 9/L)a 0.95 (0.90–0.99) 0.029
Δ Neutrophil (×10E + 9/L)a 1.31 (0.99–1.72) 0.058
Δ Fibrinogen (g/L)b 6.45 (1.31–31.69) 0.022
Δ C-reactive protein (mg/L)c 1.09 (1.01–1.18) 0.037
Δ Lactate dehydrogenase (U/L)c 1.03 (1.01–1.06) 0.007

aΔ = Max − Min.

bΔ = baseline − Min.

cΔ = Max − baseline.

Discussion

Our data indicate two baseline co-variates (age and D-dimer) on admission and four dynamic co-variates (Δs of concentrations of CRP, LDH, fibrinogen, and platelets) correlate with an increased risk of death in almost 1500 hospitalized persons with COVID-19. In our dataset, we could not confirm other co-variates such as male sex [19], and comorbidities of atherosclerotic cardiovascular disease [20] and hypertension [21] that had been excluded from the present study.

Two admission co-variates correlated with risk of death: age and D-dimer concentration. Similar correlates are reported by others in COVID-19 [8, 19, 20, 2224] and in two other coronavirus infections, severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome [25, 26]. Age related immune deficiency may be the explanation of this association but is unproved [27]. High D-dimer concentration may result from the inflammation associated with COVID-19 and subsequent activation of coagulation [28]. Several potential risk factors during hospitalization, including disseminated intra-vascular coagulation, infection, dehydration, prolonged immobilization, mechanical ventilation, and central venous catheter use may further increase D-dimer concentrations [29, 30].

Four dynamic co-variates correlated with an increased risk of death including Δs of concentrations of CRP, LDH, fibrinogen, and platelets. Similar data are rarely reported in COVID-19-infection [7, 8, 31]. Higher admission LDH concentration was reported to be a risk factor for death by different studies [19, 23, 32]. However, we found the dynamics were more predictive. Han et al. report a dynamic decrease of CRP concentration in 17 subjects with COVID-19 who recovered [31]. There are few data on dynamics of fibrinogen concentration in persons with COVID-19. Tang et al. reported differences in dynamic fibrinogen between survivor and non-survivors, but this dynamic co-variate was not identified to be a risk factor for death [7]. Admission and dynamic platelets are not reported to correlate with risk of death in persons with COVID-19 [22, 33, 34].

Our study has important limitations. It was retrospective and researchers were not blinded to the outcome when they analyzed the data. Also, we have no external validation cohort. Finally, we did not adjust for multiple comparisons. As such, our conclusions should be interpreted as exploratory and descriptive. Because subjects more likely to die have profound changes in several of these co-variates the correlations we report should not be assumed to be cause-and-effect.

In conclusion, we show admission hematological co-variates except D-dimer concentration are not associated with an increased risk of death in a large cohort of subjects with COVID-19. However, dynamic measurements of platelets, fibrinogen, CRP, and LDH correlate with risk of death. We await validation of our conclusions.

Supplementary information

Supplementary contents (130.2KB, docx)

Acknowledgements

We thank patients, families, and health care providers participating in our study. Funded by the Natural Science Foundation of China (NSFC; 81974009 to QL, 81974221 and 81470330 to ZC) and the Fundamental Research Funds for the Central Universities (2020kfyXGYJ086 to QL). RPG acknowledges support from the National Institute of Health Research Biomedical Research Center funding scheme.

Author contributions

QL, ZC, and YH designed the study. QL, YC, LeC, DW, JY, HW, WH, LC, FD, WeiC, WenC, LL, QR, QL, WR, and FG collected the data. All authors had full access to the data, were involved in data interpretation, and vouch for the accuracy of the analyses. QL, YC, LeC, DW, and RPG prepared the typescript which all authors approved final approval and supported the decision to submit for publication.

Data availability

All data generated or analyzed during this study are included in this published article and its Supplementary Information files.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

These authors contribution equally: Qiubai Li, Yulin Cao, Lei Chen, Di Wu, Jianming Yu, Hongxiang Wang, Wenjuan He, Li Chen, Fang Dong, Weiqun Chen

Contributor Information

Qiubai Li, Email: qiubaili@hust.edu.cn.

Zhichao Chen, Email: chenzhichao@hust.edu.cn.

Yu Hu, Email: dr_huyu@126.com.

Supplementary information

The online version of this article (10.1038/s41375-020-0910-1) contains supplementary material, which is available to authorized users.

References

  • 1.Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72 314 cases from the Chinese Center for Disease Control and Prevention. JAMA. 2020. 10.1001/jama.2020.2648. [DOI] [PubMed]
  • 2.Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Verity R, Okell LC, Dorigatti I, Winskill P, Whittaker C, Imai N, et al. Estimates of the severity of coronavirus disease 2019: a model-based analysis. Lancet Infect Dis. 2020;20:669–77. doi: 10.1016/S1473-3099(20)30243-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323:1061–9. doi: 10.1001/jama.2020.1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708–20. doi: 10.1056/NEJMoa2002032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:507–13. doi: 10.1016/S0140-6736(20)30211-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020;18:844–7. doi: 10.1111/jth.14768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–62. doi: 10.1016/S0140-6736(20)30566-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Zhang G, Zhang J, Wang B, Zhu X, Wang Q, Qiu S. Analysis of clinical characteristics and laboratory findings of 95 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a retrospective analysis. Respir Res. 2020;21:74. doi: 10.1186/s12931-020-01338-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin Infect Dis. 2020. 10.1093/cid/ciaa248. [DOI] [PMC free article] [PubMed]
  • 11.Liu J, Li S, Liu J, Liang B, Wang X, Wang H, et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine. 2020;55:102763. doi: 10.1016/j.ebiom.2020.102763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.He W, Chen L, Chen L, Yuan G, Fang Y, Chen W, et al. COVID-19 in persons with haematological cancers. Leukemia. 2020;34:1637–45. doi: 10.1038/s41375-020-0836-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Li W, Wang D, Guo J, Yuan G, Yang Z, Gale RP. COVID-19 in persons with chronic myeloid leukaemia. Leukemia. 2020. 10.1038/s41375-020-0853-6. [DOI] [PMC free article] [PubMed]
  • 14.WHO. Clinical management of severe acute respiratory infection when novel coronavirus (nCoV) infection is suspected: interim guidance. WHO. 2020. https://www.who.int/publications-detail/clinical-management-of-severe-acute-respiratory-infection-when-novel-coronavirus-(ncov)-infection-is-suspected. Accessed 25 Feb.
  • 15.National Health Commission of China. The novel coronavirus pneumonia diagnosis and treatment program, 7th version. China. 2020. http://www.nhc.gov.cn/yzygj/s7653p/202003/46c9294a7dfe4cef80dc7f5912eb1989.shtml. Accessed 04 Apr.
  • 16.Yang X, Yu Y, Xu J, Shu H, Xia JA, Liu H, et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med. 2020;S2213-2600:30079–5. doi: 10.1016/S2213-2600(20)30079-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhang J, Zhou L, Yang Y, Peng W, Wang W, Chen X. Therapeutic and triage strategies for 2019 novel coronavirus disease in fever clinics. Lancet Respir Med. 2020;8:e11–2. doi: 10.1016/S2213-2600(20)30071-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Gale RP. Perspective: SARS-CoV-2, COVID-19 and haematologists. Acta Haematol. 2020. 10.1159/000508021. [DOI] [PMC free article] [PubMed]
  • 19.Li X, Xu S, Yu M, Wang K, Tao Y, Zhou Y, et al. Risk factors for severity and mortality in adult COVID-19 inpatients in Wuhan. J Allergy Clin Immunol. 2020. 10.1016/j.jaci.2020.04.006. [DOI] [PMC free article] [PubMed]
  • 20.Du RH, Liang LR, Yang CQ, Wang W, Cao TZ, Li M, et al. Predictors of mortality for patients with COVID-19 pneumonia caused by SARS-CoV-2: a prospective cohort study. Eur Respir J. 2020;55:2000524. 10.1183/13993003.00524-2020. [DOI] [PMC free article] [PubMed]
  • 21.Guan WJ, Liang WH, Zhao Y, Liang HR, Chen ZS, Li YM, et al. Comorbidity and its impact on 1590 patients with Covid-19 in China: a Nationwide analysis. Eur Respir J. 2020. 10.1183/13993003.00547-2020 [DOI] [PMC free article] [PubMed]
  • 22.Tang N, Bai H, Chen X, Gong J, Li D, Sun Z, et al. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost. 2020;18:1094–9. doi: 10.1111/jth.14817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern Med. 2020. 10.1001/jamainternmed.2020.0994. [DOI] [PMC free article] [PubMed]
  • 24.Chen R, Liang W, Jiang M, Guan W, Zhan C, Wang T, et al. Risk factors of fatal outcome in hospitalized subjects with coronavirus disease 2019 From a nationwide analysis in China. Chest. 2020. 10.1016/j.chest.2020.04.010. [DOI] [PMC free article] [PubMed]
  • 25.Choi KW, Chau TN, Tsang O, Tso E, Chiu MC, Tong WL, et al. Outcomes and prognostic factors in 267 patients with severe acute respiratory syndrome in Hong Kong. Ann Intern Med. 2003;139:715–23. doi: 10.7326/0003-4819-139-9-200311040-00005. [DOI] [PubMed] [Google Scholar]
  • 26.Hong KH, Choi JP, Hong SH, Lee J, Kwon JS, Kim SM, et al. Predictors of mortality in Middle East respiratory syndrome (MERS) Thorax. 2018;73:286–9. doi: 10.1136/thoraxjnl-2016-209313. [DOI] [PubMed] [Google Scholar]
  • 27.Opal SM, Girard TD, Ely EW. The immunopathogenesis of sepsis in elderly patients. Clin Infect Dis. 2005;41:S504–12. doi: 10.1086/432007. [DOI] [PubMed] [Google Scholar]
  • 28.Connors JM, Levy JH. COVID-19 and its implications for thrombosis and anticoagulation. Blood. 2020;135:1033–45. doi: 10.1182/blood.2020006000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wang T, Chen R, Liu C, Liang W, Guan W, Tang R, et al. Attention should be paid to venous thromboembolism prophylaxis in the management of COVID-19. Lancet Haematol. 2020;7:E362–3. doi: 10.1016/S2352-3026(20)30109-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Terpos E, Ntanasis-Stathopoulos I, Elalamy I, Kastritis E, Sergentanis TN, Politou M, et al. Hematological findings and complications of COVID-19. American J Hematol. 2020. 10.1002/ajh.25829. [DOI] [PMC free article] [PubMed]
  • 31.Han X, Cao Y, Jiang N, Chen Y, Alwalid O, Zhang X, et al. Novel coronavirus pneumonia (COVID-19) progression course in 17 discharged patients: comparison of clinical and thin-section CT features during recovery. Clin Infect Dis. 2020. 10.1093/cid/ciaa271 [DOI] [PMC free article] [PubMed]
  • 32.Xie J, Hungerford D, Chen H,  Abrams S, Li S, Wang G, et al. Development and external validation of a prognostic multivariable model on admission for hospitalized patients with COVID-19. SSRN. 2020. 10.2139/ssrn.3562456.
  • 33.Xie J, Covassin N, Fan Z, Singh P, Gao W, Li G, et al. Association between hypoxemia and mortality in patients with COVID-19. Mayo Clin Proc. 2020;95:1138–47. doi: 10.1016/j.mayocp.2020.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liu Y, Du X, Chen J, Jin Y, Peng L, Wang HHX, et al. Neutrophil-to-lymphocyte ratio as an independent risk factor for mortality in hospitalized patients with COVID-19. J Infect. 2020. 10.1016/j.jinf.2020.04.002. [DOI] [PMC free article] [PubMed]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary contents (130.2KB, docx)

Data Availability Statement

All data generated or analyzed during this study are included in this published article and its Supplementary Information files.


Articles from Leukemia are provided here courtesy of Nature Publishing Group

RESOURCES