TO THE EDITOR:

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), primarily manifests as a respiratory illness and has affected >5 million people worldwide, with >350 000 deaths.1,2  There are no current approved therapies for COVID-19. Administration of convalescent plasma (CP) may be effective therapy for COVID-19.3-6  Early indicators suggest that transfusion of CP is safe in COVID-19.7  We report the early clinical experience of 20 hospitalized patients treated with CP compared with 20 matched controls with severe or life-threatening COVID-19 infection.

Twenty patients with COVID-19 infection, diagnosed using quantitative reverse-transcriptase polymerase chain reaction assay for SARS-CoV-2 on nasopharyngeal swabs, were treated with CP in 5 hospitals in the Seattle area between April 13 and April 26 of 2020. Patients with severe or critical illness were treated with 1 unit of ABO-compatible CP under an expanded access protocol (IND 19832). Baseline demographic and clinical characteristics, including comorbidities, severity of illness, laboratory parameters, and clinical outcomes, were recorded up to 14 days after CP transfusion or the equivalent day of hospitalization for controls. Donor information was also collected. This study was approved by the Institutional Review Board of Providence St. Joseph Health.

The median age of patients treated with CP was 60 years (range, 29-95), with 20% of patients older than 80 years. The most common signs and symptoms of COVID-19 illness were cough or shortness of breath (90%), lymphopenia (67%), and an abnormal radiograph (80%). The most common comorbidities were hypertension (60%), diabetes (45%), and obesity (20%). One third of patients required mechanical ventilation (MV). Median time from hospitalization to CP was early at 2 days (interquartile range [IQR], 1-4.3). The majority of patients received additional therapies, including azithromycin (60%), hydroxychloroquine (55%), or multiple combinations. CP recipients had primarily A (45%) or B (45%) ABO type.

The 8 COVID-19–recovered donors who provided units ranged in age from 29 to 79 years. All had symptoms of respiratory illness, muscle aches, and/or headache, but none required hospitalization. All were more >28 days past their last symptoms of COVID illness. Donations were well tolerated and without significant complications. Anti–SARS-CoV-2 immunoglobulin G serology, as determined by the Abbott ARCHITECT, was positive in all but 1 donor, whereas EuroImmun was negative (n = 1), equivocal (n =3), moderate positive (n = 3), or high positive (n = 1).

Figure 1 shows the clinical and laboratory parameters reported over 7 days after CP. Temperature improved in all patients after delivery of CP. The mean temperature decrease was 0.3°C in the CP group (standard deviation [SD], 0.5), whereas mean absolute lymphocyte count was 1.01 × 103/μL (SD, 0.47) and did not change by day 7 (1.09 × 103/μL; SD, 0.55). The mean baseline C-reactive protein was 170.5 mg/L (SD, 137.6), which decreased to 127.5 mg/L (SD, 144.0) after CP. A decrease in the fraction of inspired oxygen (FiO2) from 53% (SD, 29) to 47% (SD, 24) was seen by day 7 after CP. For patients who were on MV, the mean Pao2/FiO2 of inspired oxygen ratio at baseline was 81 (SD, 23), which improved to 96 (SD, 22). Two patients were liberated from the ventilator, whereas 2 required intubation after CP.

Figure 1.

Serial change in laboratory and clinical parameters in CP and control patients. Solid lines represent a smoothing spline through the observed data points. CRP, C-reactive protein; PEEP, positive end-expiratory pressure; P/F, Pao2/FiO2.

Figure 1.

Serial change in laboratory and clinical parameters in CP and control patients. Solid lines represent a smoothing spline through the observed data points. CRP, C-reactive protein; PEEP, positive end-expiratory pressure; P/F, Pao2/FiO2.

Close modal

The outcomes of all patients are reported in Table 1. Median World Health Organization (WHO) ordinal scale score was 5 at CP infusion, which improved to 4.5 at day 7 and 3.5 at day 14. No adverse events with CP were reported. The incidence of venous thromboembolism (VTE) was (20%). At 7 days of follow-up, 25% of patients were discharged, whereas 10% had died. The 2 deaths occurred in patients who had been intubated for >2 weeks each and chose to transition to comfort measures. No patients died if they received CP prior to 7 days of hospitalization. No additional deaths occurred in the CP group by day 14.

Table 1.

Clinical outcomes

Clinical statusCP (n = 20)Matched controls (n = 20)
Baseline (d 0)Status at 7-d follow-upStatus at 14-d follow-upBaseline*Status at 7-d follow-upStatus at 14-d follow-up
WHO ordinal scale score       
 Median (IQR) 5 (4.0-6.3) 4.5 (4.0-6.0) 3.5 (0-6) 5 (4-7) 4.5 (0.8-7.3) 3 (0-8) 
 Mean (SD) 5.2 (1.3) 5.07 (1.4) 3.1 (3.1) 5.4 (1.4) 4.2 (3.2) 3.45 (3.6) 
VTE 4 (20) 4 (20) 4 (20) 4 (20) 4 (20) 4 (20) 
ICU       
 MV 6 (30) 6 (30) 4 (20) 6 (30) 5 (25) 1 (5) 
 Duration of MV (IQR), d 14.5 (6-17.8) 11.5 (7.5-9.8) 16.5 (12.8-20) 3 (2.3-3.8) 9 (7-0) 11 (8.5-13.5) 
Extubated survivors — 1 (5) 2 (10) — 0 (0) 3 (15) 
Adverse event after CP 0 (0) 0 (0) 0 (0) — — — 
Hospital stay, d       
 LOS prior to CP, median (range) 2 (1-21) — — — — — 
 LOS prior to CP, median (IQR) 2 (1-4.3) — — — — — 
 Total LOS, median (IQR) — 9 (8-11.3) 15 (10-16.5) — 8 (7-9.5) 9 (7-13.5) 
 LOS of nonsurvivors, median (IQR) — 19.5 (15.8-23.3) 19.5 (15.8-23.3) — 8 (7-9) 8.5 (7.3-10.5) 
 LOS of survivors, median (IQR) — 9 (8-10) 15 (10-16) — 8 (7-9.5) 10 (7.3-15) 
Discharged — 5 (25) 9 (45) — 7 (35) 9 (45) 
Deaths — 2 (10) 2 (10) — 5 (25) 6 (30) 
 CP given prior to 7 d of hospitalization — 0 (0) 0 (0) — 4 (20) 5 (25) 
 CP given after 7 d of hospitalization — 2 (10) 2 (10) — 1 (5) 1 (5) 
Clinical statusCP (n = 20)Matched controls (n = 20)
Baseline (d 0)Status at 7-d follow-upStatus at 14-d follow-upBaseline*Status at 7-d follow-upStatus at 14-d follow-up
WHO ordinal scale score       
 Median (IQR) 5 (4.0-6.3) 4.5 (4.0-6.0) 3.5 (0-6) 5 (4-7) 4.5 (0.8-7.3) 3 (0-8) 
 Mean (SD) 5.2 (1.3) 5.07 (1.4) 3.1 (3.1) 5.4 (1.4) 4.2 (3.2) 3.45 (3.6) 
VTE 4 (20) 4 (20) 4 (20) 4 (20) 4 (20) 4 (20) 
ICU       
 MV 6 (30) 6 (30) 4 (20) 6 (30) 5 (25) 1 (5) 
 Duration of MV (IQR), d 14.5 (6-17.8) 11.5 (7.5-9.8) 16.5 (12.8-20) 3 (2.3-3.8) 9 (7-0) 11 (8.5-13.5) 
Extubated survivors — 1 (5) 2 (10) — 0 (0) 3 (15) 
Adverse event after CP 0 (0) 0 (0) 0 (0) — — — 
Hospital stay, d       
 LOS prior to CP, median (range) 2 (1-21) — — — — — 
 LOS prior to CP, median (IQR) 2 (1-4.3) — — — — — 
 Total LOS, median (IQR) — 9 (8-11.3) 15 (10-16.5) — 8 (7-9.5) 9 (7-13.5) 
 LOS of nonsurvivors, median (IQR) — 19.5 (15.8-23.3) 19.5 (15.8-23.3) — 8 (7-9) 8.5 (7.3-10.5) 
 LOS of survivors, median (IQR) — 9 (8-10) 15 (10-16) — 8 (7-9.5) 10 (7.3-15) 
Discharged — 5 (25) 9 (45) — 7 (35) 9 (45) 
Deaths — 2 (10) 2 (10) — 5 (25) 6 (30) 
 CP given prior to 7 d of hospitalization — 0 (0) 0 (0) — 4 (20) 5 (25) 
 CP given after 7 d of hospitalization — 2 (10) 2 (10) — 1 (5) 1 (5) 

Unless otherwise noted, all data are n (%).

LOS, length of stay; —, not applicable.

*

Baseline day 0 for matched controls corresponds to equivalent hospital day as CP transfusion.

Control patients (n = 20) were well matched with regard to age, number of comorbidities, WHO score, sequential organ failure assessment score, and severity of illness. The median WHO score improved from 5 to 4.5 at day 7 and 3 at day 14. Half of the control patients received remdesivir (RDV). VTE incidence was 20%. After 7 days of follow-up, 35% of the controls were discharged, whereas 25% had died. One additional death occurred among the controls by day 14.

We report our experience using CP in the treatment of 20 severely and critically ill hospitalized COVID-19 patients and 20 matched controls. Although laboratory and respiratory parameters were improved in patients following CP infusion, their status was similar to that of controls. A similar proportion of patients in each group was discharged, whereas the 7- and 14- day fatality rate in CP patients compared favorably to that in controls. CP infusion was safe without adverse events. There was no evidence of clinical worsening to suggest a hyperimmune response.8  We did not see an increased risk for VTE in CP patients, although the incidence was high in both groups, despite heparin prophylaxis, as seen in COVID-19.9-11 

In recent publications describing outcomes for hospitalized COVID-19 patients, mortality ranged between 26% and 88%.12-18  No deaths occurred in patients who received CP within 7 days of hospitalization, with the majority of patients receiving CP early. Findings from a prior study of patients with SARS-CoV-1 reported a survival benefit with early administration of CP (prior to 14 days after symptoms), as did a meta-analysis of studies conducted during Spanish influenza.19,20  These findings, along with a recent study reporting a lack of benefit when CP is administered later (median of 21.5 days after diagnosis) for SARS-CoV-2, support that earlier treatment may be of critical importance.21  Because viral loads peak in the first week for most viral infections, deterioration in the following weeks is thought to result from inflammatory destruction of lung tissue and would not be expected to improve with CP.19 

Other than time to infusion, it is not clear how the volume of CP or neutralizing antibody titers of donors and/or recipients affect outcomes. In a prior study of CP for SARS-CoV-1, antibody titers and CP volumes did not appear to correlate with clinical response; however, patients who were positive via a quantitative reverse-transcriptase polymerase chain reaction assay and seronegative at the time of CP had a better outcome than did those who were already seropositive (66.7% vs 20%; P = .001).19  In 3 case reports of CP in SARS-CoV-2, viral loads became negative soon after CP, suggesting that antibodies from CP may contribute to viral clearance.5,6,21  However, additional factors in CP may be contributing to clinical improvement, because 4 of 10 patients reported by Duan et al had high neutralizing titers of 1:640 prior to CP.5 

The limitations of this study included small sample size and short follow-up. Also, patients treated with CP and controls received additional therapies for COVID-19. The disparity in the use of RDV between the 2 groups is stark, with the CP group at 5% and controls at 50%. There are emerging data from the National Institutes of Health that RDV may result in a 31% improvement in time to recovery.22-24  If this effect was present in our control patients, then the outcome with CP may be even more remarkable.

In conclusion, the current study suggests that CP use in severe and critically ill patients with COVID-19 may improve survival if given early in the course of disease. The efficacy as a potential therapy needs further study in well-designed trials to better understand the contribution of CP to outcomes in COVID-19.

Data sharing requests should be sent to Livia Hegerova (livia.hegerova@swedish.org).

The authors thank all patients and their families for participating in the study. They also thank Mark H. Wener and Anu Chaudhary for helping to facilitate serologic testing.

This work was supported in part by research funding from Barbara and Kent Chaplin, a US Department of Health and Human Services (HHS), Biomedical Advanced Research and Development Authority (BARDA) grant contract 75A50120C00096, National Institutes of Health, National Center for Advancing Translational Seciences (NCATS) grant UL1TR002377, Schwab Charitable Fund (Eric E Schmidt, Wendy Schimdt donors), United Health Group, National Basketball Association (NBA), Millennium Pharmaceuticals, Octopharma Octapharma USA, Inc, and the Mayo Clinic.

Contribution: L.H., N.B., M.B., V.D., K.P., J.M.P., T.A.G., K.A.S., and C.M. reviewed the literature, enrolled patients in the study, collected, analyzed, and interpreted data, and wrote the manuscript; K.A., R.H., J.M.J., and B.A.K. recruited donors and analyzed data; A.C.L. and M.L.A. collected data; J.D.G. was a member of the research advisory committee; and J.S.P., A.L., S.-j.L., and M.D.S. contributed control patient material.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Livia Hegerova, Center for Blood Disorders and Stem Cell Transplantation, Swedish Cancer Institute, 1221 Madison St, Suite 1000, Seattle, WA 98104; e-mail: livia.hegerova@swedish.org.

1.
World Health Organization
. Coronavirus disease (COVID-19) pandemic. Available at: https://www.who.int/emergencies/diseases/novel-coronavirus-2019. Accessed 25 May 2020.
2.
Dong
E
,
Du
H
,
Gardner
L
.
An interactive web-based dashboard to track COVID-19 in real time
.
Lancet Infect Dis
.
2020
;
20
(
5
):
533
-
534
.
3.
Zhang
B
,
Liu
S
,
Tan
T
, et al
.
Treatment with convalescent plasma for critically ill patients with severe acute respiratory syndrome coronavirus 2 infection [published online ahead of print 31 March 2020]
.
Chest
. doi:10.1016/j.chest.2020.03.039.
4.
Roback
JD
,
Guarner
J
.
Convalescent plasma to treat COVID-19: possibilities and challenges
.
JAMA
.
2020
;
323
(
16
):
1561
.
5.
Duan
K
,
Liu
B
,
Li
C
, et al
.
Effectiveness of convalescent plasma therapy in severe COVID-19 patients
.
Proc Natl Acad Sci USA
.
2020
;
117
(
17
):
9490
-
9496
.
6.
Shen
C
,
Wang
Z
,
Zhao
F
, et al
.
Treatment of 5 critically ill patients with COVID-19 with convalescent plasma
.
JAMA
.
2020
;
323
(
16
):
1582
.
7.
Joyner
M
,
Wright
RS
,
Fairweather
D
, et al
.
Early safety indicators of COVID-19 convalescent plasma in 5,000 patients [published online ahead of print 11 June 2020]
.
J Clin Invest
. doi:10.1101/2020.05.12.20099879.
8.
Wang
SF
,
Tseng
SP
,
Yen
CH
, et al
.
Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins
.
Biochem Biophys Res Commun
.
2014
;
451
(
2
):
208
-
214
.
9.
Klok
FA
,
Kruip
M
,
van der Meer
NJM
, et al
.
Incidence of thrombotic complications in critically ill ICU patients with COVID-19
.
Thromb Res
.
2020
;
191
:
145
-
147
.
10.
Yin
S
,
Huang
M
,
Li
D
,
Tang
N
.
Difference of coagulation features between severe pneumonia induced by SARS-CoV2 and non-SARS-CoV2 [published online ahead of print 3 April 2020]
.
J Thromb Thrombolysis
. doi:10.1007/s11239-020-02105-8.
11.
Thachil
J
,
Tang
N
,
Gando
S
, et al
.
ISTH interim guidance on recognition and management of coagulopathy in COVID-19
.
J Thromb Haemost
.
2020
;
18
(
5
):
1023
-
1026
.
12.
Bhatraju
PK
,
Ghassemieh
BJ
,
Nichols
M
, et al
.
Covid-19 in critically ill patients in the Seattle region - case series
.
N Engl J Med
.
2020
;
382
(
21
):
2012
-
2022
.
13.
Guan
WJ
,
Ni
ZY
,
Hu
Y
, et al;
China Medical Treatment Expert Group for COVID-19
.
Clinical characteristics of coronavirus disease 2019 in China
.
N Engl J Med
.
2020
;
382
(
18
):
1708
-
1720
.
14.
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
;
323
(
13
):
1239
.
15.
Yang
X
,
Yu
Y
,
Xu
J
, et al
.
Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study
.
Lancet Respir Med
.
2020
;
8
(
5
):
475
-
481
.
16.
Zhou
F
,
Yu
T
,
Du
R
, et al
.
Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study
.
Lancet
.
2020
;
395
(
10229
):
1054
-
1062
.
17.
Arentz
M
,
Yim
E
,
Klaff
L
, et al
.
Characteristics and outcomes of 21 critically ill patients with COVID-19 in Washington state
.
JAMA
.
2020
;
323
(
16
):
1612
.
18.
Richardson
S
,
Hirsch
JS
,
Narasimhan
M
, et al;
the Northwell COVID-19 Research Consortium
.
Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area
.
JAMA
.
2020
;
323
(
20
):
2052
.
19.
Cheng
Y
,
Wong
R
,
Soo
YO
, et al
.
Use of convalescent plasma therapy in SARS patients in Hong Kong
.
Eur J Clin Microbiol Infect Dis
.
2005
;
24
(
1
):
44
-
46
.
20.
Luke
TC
,
Kilbane
EM
,
Jackson
JL
,
Hoffman
SL
.
Meta-analysis: convalescent blood products for Spanish influenza pneumonia: a future H5N1 treatment?
Ann Intern Med
.
2006
;
145
(
8
):
599
-
609
.
21.
Zeng
QL
,
Yu
ZJ
,
Gou
JJ
, et al
.
Effect of convalescent plasma therapy on viral shedding and survival in patients with coronavirus disease 2019
.
J Infect Dis
.
2020
;
222
(
1
):
38
-
43
.
22.
Wang
Y
,
Zhang
D
,
Du
G
, et al
.
Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial
.
Lancet
.
2020
;
395
(
10236
):
1569
-
1578
.
23.
National Institutes of Health
.
NIH clinical trial shows remdesivir accelerates recovery from advanced COVID-19
. Available at: https://www.nih.gov/news-events/news-releases/nih-clinical-trial-shows-remdesivir-accelerates-recovery-advanced-covid-19. Accessed 29 April 2020.
24.
Beigel
JH
,
Tomashek
KM
,
Dodd
LE
, et al;
ACTT-1 Study Group Members
.
Remdesivir for the treatment of Covid-19 - preliminary report [published online ahead of print 22 May 2020]
.
N Engl J Med
. doi:10.1056/NEJMoa2007764.
Sign in via your Institution