ArticlePDF Available

Association of soluble PD-L1 and NLR combination with 1-Year mortality in patients with COVID-19

Authors:

Abstract

Purpose Understanding the relationship between patient immune characteristics, disease severity, and mortality represents a critical step in the fight against COVID-19. Elevated levels of programmed death ligand-1 (PD-L1) and Neutrophil-lymphocyte ratio (NLR) are linked to increased severity of acute COVID-19 in patients. This study aimed to investigate the association of the combination of sPD-L1 and NLR with 1-year Mortality in patients with COVID-19. Methods A prospective study was conducted involving patients with COVID-19 in Karaganda, Kazakhstan. The level of sPD-L1 in the blood serum was evaluated by ELISA. The effect of biomarkers on the development of mortality was analyzed with multivariate regression. Results The risk of mortality within one year HR was 2.46 if the plasma sPD-L1 value of more than 277.13 pg/ml, and for NLR more than 2.46 HR was 2.87. The model of combining sPD-L1 and NLR resulted in an improvement in the predictive accuracy of the Hazard Ratio 7.6 (95 % CI: 3.02–19.11). Conclusion The combination of two immune-mediated markers (sPD-L1 and NLR), which reflect the systemic inflammatory balance of activation and exhaustion, can complement each other and improve the assessment of the risk of death in patients with COVID-19.
International Immunopharmacology 129 (2024) 111600
Available online 6 February 2024
1567-5769/© 2024 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
Association of soluble PD-L1 and NLR combination with 1-Year mortality in
patients with COVID-19
Lyudmila Akhmaltdinova
a
, Irina Mekhantseva
a
,
*
, Lyudmila Turgunova
a
, Mikhail Kostinov
b
,
Zhibek Zhumadilova
a
, Anar Turmukhambetova
a
a
Karaganda Medical University, Scientic and Research Center, Karaganda, Kazakhstan
b
I.I. Mechnikov Research Institute of Vaccines and Sera, Sechenov First Moscow State Medical University, Department of Epidemiology and Modern Vaccination
Technologies, Moscow, Russia
ARTICLE INFO
Keywords:
COVID-19
Post-COVID Mortality
sPDL-1
Neutrophil-to-Lymphocyte Ratio
Humoral and cellular immune response
ABSTRACT
Purpose: Understanding the relationship between patient immune characteristics, disease severity, and mortality
represents a critical step in the ght against COVID-19. Elevated levels of programmed death ligand-1 (PD-L1)
and Neutrophil-lymphocyte ratio (NLR) are linked to increased severity of acute COVID-19 in patients. This study
aimed to investigate the association of the combination of sPD-L1 and NLR with 1-year Mortality in patients with
COVID-19.
Methods: A prospective study was conducted involving patients with COVID-19 in Karaganda, Kazakhstan. The
level of sPD-L1 in the blood serum was evaluated by ELISA. The effect of biomarkers on the development of
mortality was analyzed with multivariate regression.
Results: The risk of mortality within one year HR was 2.46 if the plasma sPD-L1 value of more than 277.13 pg/ml,
and for NLR more than 2.46 HR was 2.87. The model of combining sPD-L1 and NLR resulted in an improvement
in the predictive accuracy of the Hazard Ratio 7.6 (95 % CI: 3.0219.11).
Conclusion: The combination of two immune-mediated markers (sPD-L1 and NLR), which reect the systemic
inammatory balance of activation and exhaustion, can complement each other and improve the assessment of
the risk of death in patients with COVID-19.
1. Introduction
SARS-CoV-2 has had a signicant impact on public health and has
forced a re-evaluation of traditional epidemic management approaches.
Data from multiple epidemiological studies indicate that individuals
previously infected with COVID-19 are at a signicantly higher risk of
death within the next 12 months [1,2]. Predictors of COVID-19 severity
and mortality in the acute period are well-studied; long-term conse-
quences, including mortality, are still under investigation. Understand-
ing the relationship between patient immune characteristics, disease
severity, and mortality represents a critical step in the ght against
COVID-19. Severe and critical manifestations of COVID-19 are caused by
a dysregulated immune response, in which the adaptive immune system
driven by T and B lymphocytes plays a fundamental role [3].
Previous studies have established the importance of elevated levels
of programmed death ligand-1 (PD-L1) and its receptor PD-1 in the
pathogenesis of cancer [47] and various infectious diseases, including
AIDS and hepatitis [810] There is evidence to suggest that elevated
levels of PD-L1 might be linked to increased severity of acute COVID-19
in patients [11,12]. The interaction between the checkpoint molecules
PD-1 and its ligand PD-L1 helps to reduce inammation and tissue
damage in severe and critical cases of COVID-19 [13,14]. However, due
to increased levels of PD-1 expression, T cells can become exhausted and
their effector functions may decrease, leading to the progression of the
disease. Beserra et al. analyzed the levels of sPD-1 and sPD-L1 in the
serum of patients and observed an increase in sPD-L1 in patients with
severe and critical symptoms [15]. Additionally, it has been reported
that increased levels of sPD-L1 were associated with lower lymphocyte
counts and higher CRP levels, and were also linked to longer hospital
stays and higher in-hospital mortality rates [16]. However, the mecha-
nisms underlying increased PD-L1 levels in SARS-CoV-2 are not fully
understood, and studies on soluble PD-L1 in COVID-19 patients with
* Corresponding author.
E-mail addresses: immunol.lab@gmail.com (L. Akhmaltdinova), mehanceva@qmu.kz (I. Mekhantseva), turgunova@qmu.kz (L. Turgunova), monolit.96@mail.ru
(M. Kostinov), Zhumadilova.Z@qmu.kz (Z. Zhumadilova), turmuhambetova@qmu.kz (A. Turmukhambetova).
Contents lists available at ScienceDirect
International Immunopharmacology
journal homepage: www.elsevier.com/locate/intimp
https://doi.org/10.1016/j.intimp.2024.111600
Received 12 December 2023; Received in revised form 16 January 2024; Accepted 24 January 2024
International Immunopharmacology 129 (2024) 111600
2
varying disease severity are scarce.
It has been established that systemic inammation is a key factor in
COVID-19 development [1719]. Baseline Neutrophil-lymphocyte ratio
(NLR) level has been proposed as a prognostic marker in COVID-19 [20].
Compelling evidence of immune dysfunction associated with COVID-
19 and its long-term impact on mortality is essential for effective patient
stratication in planning preventive measures and clinical management
in the post-covid period. Therefore, this study aimed to investigate the
association of the combination of immune-mediated mediators such as
sPD-L1 and NLR with 1-year Mortality in patients with COVID-19.
2. Materials and Methods
2.1. Study design and participants
A prospective study was conducted at the infectious diseases clinics
of Karaganda Regional Clinical Hospital and Karaganda Medical Uni-
versity Hospital between May and August 2021. The main group for this
study included patients over 18 years of age who had tested positive for
COVID-19. Pregnant and lactating women, and individuals with
compromised immune systems, such as those affected by human im-
munodeciency virus (HIV) infection, or those currently undergoing
treatment for malignancies, were excluded from the study. The study
involved 314 COVID-19-positive patients conrmed by RT-PCR analysis
of nasopharyngeal swabs for genomic RNA with or without co-existing
conditions, undergoing treatment at an infectious diseases hospital.
Clinical data and laboratory results were collected from patients
electronic medical records. All laboratory tests, including complete
blood count, plasma C-reactive protein (CRP), ferritin, and D-dimer
concentrations, were collected on the same day as plasma sampling.
Clinical outcomes (intensive care unit admission, length of hospital stay,
mortality) were recorded until hospital discharge. To assess the severity
of COVID-19 in patients, WHO criteria were used [21]. Two groups of
patients were identied: the rst group included those with moderate
severity, while the second group consisted of patients with severe and
critical disease severity. Electronic medical records were used to collect
data on the disease progression, comorbidities, anthropometric in-
dicators, heart rate (HR), oxygen saturation, and percentage of lung
tissue damage. Body mass index and Charlson comorbidity index were
calculated for all patients [22]. All examinations were carried out on the
rst day of admission to the hospital. After discharge, patients were
monitored via phone and recorded in the medical information system for
treatment and death. The endpoint considered all deaths, including
cause and date. The observation period for COVID-19 patients was one
year from hospital admission, with survivors dened as those alive after
this time.
2.2. Laboratory analysis
Blood samples were collected on admission day by venipuncture into
2 vacuum tubes containing 5 ml EDTA. Plasma was collected and ali-
quoted, and serum aliquots were stored at 80 C. Enzyme-linked
immunosorbent assays (ELISA) were used to quantify plasma sPD-L1
concentrations with a research-only reagent kit (Abcam, #ab277712).
Colourimetric determinations were recalculated using a four-parameter
logistic curve corresponding to each plate. A complete blood count was
performed on a hematology analyzer (Mindray, China).
2.3. Statistical analysis
The data from the study was analyzed statistically using SPSS version
21.0. (IBM Corp., Armonk, NY, USA) и MedCalc version 22 (MedCalc
Software Ltd, Ostland, Belgium). Data presented in tables and graphs
using GraphPad Prism software version 9.5 (GraphPad Software, Inc.,
San Diego, California, USA). The normality of the distribution was
assessed using the Kolmogorov-Smirnov test. Quantitative measures
with non-normal distribution are described by median (Me) and inter-
quartile range. Qualitative characteristics are described with percent-
ages. Comparative analysis of quantitative data between groups utilized
the Mann-Whitney score for nonparametric distributions and Fishers
exact tests for categorical variables. The discrimination score was
determined by the area under the curve (AUC) of the receiver operating
characteristic (ROC). AUCs with 95 % condence intervals were calcu-
lated to estimate the diagnostic value of NLR and sPD-L1. Optimal cut-
off values for these biomarkers were determined based on the
maximum Youdens J index and further used to estimate survival
considering high and low biomarker concentrations using Kaplan-Meier
survival analysis. Hazard ratios were based on the log-rank (Mantel-Cox)
test. Using multivariate regression, the effect of biomarkers on the
development of mortality was analyzed, adjusting for already proven
risk variables such as gender, age, comorbidity, and disease severity.
Statistical test differences were considered signicant if p values were <
0.05.
2.4. Ethical declare
This study was conducted in accordance with the Declaration of
Helsinki and approved by the Bioethics Committee of Karaganda Med-
ical University No. 18, dated 14 April 2021. Written informed consent
was obtained from the participants.
3. Results
3.1. Demographic data and clinical and laboratory characteristics of
patients
The study population (n =314) was divided into groups based on
disease severity (moderate disease [n =245] vs. severe disease [n =69]:
49 severe patients (71 %) and 20 patients (29 %) with critical disease
severity) and outcome (survivors [n =275] vs. non-survivors [n =39]).
Table 1 displays patient characteristics, grouped by disease severity and
mortality. The patients who contracted COVID-19 had an average age of
63 years. There was no signicant difference in the median age between
patients with moderate and severe severity. However, the median age of
those who died was 72 years, while the median age of survivors was 61
years (p =0.0001). Patients with severe disease and those who died had
a higher prevalence of comorbidities. Out of the 184 patients (58.5 %)
had chronic diseases, including hypertension (57.6 %), diabetes mellitus
(26.1 %), cardiovascular diseases (42 %), chronic pulmonary diseases
(2.5 %), and chronic kidney disease (4.7 %). The group of patients with
severe COVID-19 and the group of deceased patients had higher levels of
leukocytes, neutrophils, NLR, and ESR, as well as inammatory markers
such as CRP, ferritin, and D-dimer (Table 1). The median hospitalization
time for patients with COVID-19 differed between the moderate and
severe disease groups (p =0.0001). Moreover, patients with severe
illness and those who unfortunately passed away had a higher chance of
receiving respiratory assistance and being admitted to the intensive care
unit (ICU) (p =0.0001). Comparable ndings were observed while
comparing the groups of survived and deceased patients (p =0.0001).
3.2. sPD-L1 concentrations in patients with COVID-19
Severe COVID-19 patients have been found to have higher plasma
concentrations of sPD-L1 NLR levels compared to patients with moder-
ate disease. Fig. 1A shows that sPD-L1 levels were 279.1 [183.4401.3]
pg/mL for severe patients and 225.9 [150.7329.5] pg/mL for moderate
patients, indicating a statistically signicant difference (p =0.011).
Similarly, Fig. 1B shows that the NLR levels were 3.4 [2.45.3] pg/ml for
severe patients and 2.4 [1.53.5] pg/mL for moderate patients (p =
0.0001). Similarly, non-survivors had higher plasma sPD-L1 concen-
trations (Fig. 1C: 295.6 [177.8390.4] pg/ml vs 228.6 [154.4336.2]
pg/ml, p =0.045) and NLR (Fig. 2D: 3.4 [2.48.4] pg/ml vs. 2.4
L. Akhmaltdinova et al.
International Immunopharmacology 129 (2024) 111600
3
[1.63.7] pg/ml, p =0.0001) compared with survivors. Additionally,
the relationship between markers and disease severity was separately
assessed in subgroups of survivors and deceased to provide evidence that
plasma concentrations of sPD-L1 and NLR are associated with mortality,
independent of disease severity (Supplementary Fig. A1). In the
deceased group, there were no statistically signicant differences in the
concentrations of sPD-L1 and NLR in patients with moderate and severe
COVID-19.
3.3. Discriminatory impact of sPD-L1 and NLR integration on mortality
The diagnostic value of sPD-L1 and NLR in patients with COVID-19
was assessed using ROC curves. To assess discrimination between sur-
vivors and deceased based on concentrations, the results are presented
as ROC curves (Fig. 2). Table 2 presents the characteristics of the ROC
analyses. The calculated AUC for sPD-L1 was 0.609 (95 % CI:
0.5520.663, p =0.033), indicating moderate discrimination between
survivors and deceased. The discriminatory power of NLR was slightly
higher and also statistically signicant (AUC =0.684, 95 % CI:
0.6420.723; p =0.0001). Threshold values for these markers were
based on the maximum Youdens J index obtained from the ROC curves.
Survival of patients was stratied based on these markers. Hazard ratios
were calculated using the log-rank test (Mantel-Cox). The study found
that patients with plasma sPD-L1 levels greater than 277.13 pg/mL had
a higher risk of mortality within a year (HR =2.46, 95 % CI: 1.274.74,
shown in Fig. 3A). Similarly, patients with a high NLR greater than 2.46
(shown in Fig. 3B) had a hazard ratio for mortality of 2.87 (95 % CI:
1.495.53; p =0.0015). When creating the model, combining the cutoff
values of sPD-L1 and NLR, the Hazard Ratio was 7.6 (95 % CI:
3.0219.11, Fig. 3C), thereby proving a synergistic relationship between
sPD-L1 and NLR in deceased.
3.4. Multivariate regression analysis to predict mortality in patients with
COVID-19
A multivariate regression model was developed to forecast the pos-
sibility of death in patients admitted to the hospital with COVID-19. For
this model, patients were classied with high or low levels of sPD-L1 and
NLR according to cutoff values (Table 2). The model that included high
levels of sPD-L1 and NLR demonstrated a high predictive variable (OR
2.359, p =0.044), even after adjusting the regression model by gender
(OR 1.334, p =0.480), age over 60 years (OR 4.183, p =0.006),
Charlson Comorbidity Index (OR 2.489, p =0.147) and severity of
illness (OR 1.990, p =0.108) (Table 3).
4. Discussion
Our study ndings provide evidence to support the hypothesis that
sPD-L1 levels are increased in patients with severe COVID-19 and those
who have died. These results indicate that sPD-L1 may contribute to the
progression of COVID-19 and could be a potential target for further
immunotherapy.
Table 1
Demographic and clinical characteristics of COVID-19 patients.
All patients
(n =314)
Moderate illness
(n =245)
Severe illness
(n =69)
p-
value*
Survivors (n =275) Non-survivors (n =
39)
p-
value**
Demographic
Age (year) 63 (5171) 61 (5071) 67 (5672) 0.082 61 (4970) 72 (6382) 0.0001
Sex, n (%)
Male
Female
121 (38.6)
193 (61.4)
97 (39.6)
148 (60.4)
24 (34.8)
45 (65.2)
0.469 170 (61.8)
105 (38.2)
16 (41)
23 (59)
0.733
BMI (kg/m
2
) 29.4 (25.434.1) 28.9 (25.433.6) 30.8 (25.735.5) 0.152 29.4 (25.534.2) 28.8 (23.633.9) 0.368
Comorbidities or coexisting disorders, n (%)
Hypertension 181 (57.6) 134 (54.6) 47 (68.1) 0.046 158 (57.4) 23 (58.9) 0.857
Cardiovascular disease 132 (42) 92 (37.5) 40 (57.9) 0.002 112 (40.7) 20 (51.2) 0.211
Diabetes mellitus 82 (26.1) 60 (24.4) 22 (31.8) 0.217 72 (26.1) 10 (25.6) 0.943
Pulmonary disease 8 (2.5) 7 (2.8) 1 (1.4) 0.883 6 (2.18) 2 (5.1) 0.443
Chronic renal failure 15 (4.7) 6 (2.4) 9 (13.01) 0.002 13 (4.7) 2 (5.1) 0.913
Charlson Comorbidity Index, grade
01 201 (64) 166 (68) 35 (51) 0.015 183 (67) 18 (46) 0.002
23 85 (27) 61 (25) 24 (35) 0.015 74 (27) 11 (28) 0.002
>4 28 (9) 18 (7) 10 (14) 0.015 18 (6) 10 (26) 0.002
Vital signs at day of sampling, n (%)
Heart rate (bpm) 80 (7687) 80 (7686) 82 (7690) 0.248 80 (7686) 80 (7690) 0.516
Temperature (C) 36.7 (36.537.2) 36.7 (36.537.2) 36.7 (36.637.3) 0.337 36.7 (36.537.2) 36.8 (36.637.5) 0.128
Respiratory rate (vpm) 19 (1821) 19 (1820) 20 (1822) 0.004 19 (1820) 22 (1922) 0.0001
Oxygen saturation, % 95 (9398) 96 (9498) 93 (9096) 0.0001 95 (9498) 95 (9198) 0.116
Inltrate on chest radiograph,
%
25 (1240) 20 (832) 56 (4566) 0.0001 25 (1240) 40 (2052) 0.012
Hospital length of stay (days) 10 (912) 10 (811) 11 (915) 0.0001 10 (911) 11 (815) 0.383
Invasive mechanical
ventilation
20 (6.3) 20 (29) 8 (2.9) 12 (30.8) 0.0001
Mortality 39 (12.4) 22 (9) 17 (24.6) 0.0001 NA NA NA
Laboratory ndings
Leukocytes x 10
9
/L 5.2 (3.96.5) 5.1 (3.96.2) 5.8 (4.27.0) 0.027 5.2 (3.96.3) 5.9 (4.57.2) 0.043
Neutrophils x 10
9
/L 3.29 (2.304.49) 3.17 (2.204.34) 3.95 (2.815.47) 0.001 3.24 (2.244.36) 4.07 (2.915.58) 0.004
NLR 2.6 (1,63.8) 2.4 (1,53.5) 3.4 (2.45.3) 0.0001 2.4 (1.63.7) 3.4 (2.48.4) 0.0001
ESR, (mm/h) 15 (1022) 15 (1023) 15 (1020) 0.0001 15 (1022) 15 (820) 0.597
CRP (mg/l) 15.1 (6.174.7) 12.0 (6.037.3) 35.9 (12.0145.2) 0.0001 12.0 (6.046.4) 47.1 (8.2104.8) 0.031
Ferritin (
μ
g/l) 238 (148375) 233 (130356) 254 (186440) 0.039 234 (148361) 244 (163435) 0.335
D-dimer, (ng/ml) 301 (181441) 286 (165423) 378 (205523) 0.004 286 (169425) 425 (305549) 0.0001
sPD-L1 (pg/ml) 232.5
(159.1344.5)
225.9
(150.7329.5)
279.1
(183.4401.3)
0.011 228.6
(154.4336.2)
295.6 (177.8390.4) 0.045
Data are presented as median with interquartile ranges or n (%). Patient data were compared using the chi-square test, or Fishers exact test for categorical variables
and one-way analysis of variance. MannWhitney, nonparametric t-test was used for continuous variables. p <0.05 was considered statistically signicant. *Moderate
versus severe illness. **Survivors versus non-survivors. Abbreviations: Body mass index (BMI), Blood pressure (BP), Intensive care unit (ICU), C-reactive protein (CRP),
Erythrocyte sedimentation rate (ESR).
L. Akhmaltdinova et al.
International Immunopharmacology 129 (2024) 111600
4
Fig. 1. The plasma concentration of sPD-L1 (A) and NLR (B) moderate and severely ill patients, survivor, and non-survivor patients (C and D). Data are presented as
median with interquartile ranges, p-values were calculated with MannWhitney U tests.
Fig. 2. Receiver operating characteristic (ROC) curves of sPD-L1 (A) and NLR (B) concentrations to predict mortality among patients with COVID-19. The area under
the curve (AUC) and the p-values for signicant are depicted in the graphic.
L. Akhmaltdinova et al.
International Immunopharmacology 129 (2024) 111600
5
Multiple retrospective studies have shown that uncontrolled immune
responses and hyperinammation are hallmarks of severe COVID-19
[23,24]. The SARS-CoV-2 virus leads to a decrease in the number of
lymphocytes, additionally, it can also cause the shrinking of secondary
lymphoid organs, such as the spleen and lymph nodes [25,26]. Bio-
markers of SARS-CoV-2 infection may include overproduction of cyto-
kines that can cause lung damage, such as interleukin-1β (IL-6β), IL-2,
interferon-γ (IFN-γ), tumor necrosis factor
α
(TNF-
α
) or transforming
growth factor β (TGF-β) [2730]. CD4 and CD8 T cells in COVID-19
patients express markers of T cell exhaustion, including PD-1 and Tim-
3, which contribute to SARS-CoV-2-induced sepsis and death [13].
The main hypothesis for the immunopathogenesis of long-term COVID-
associated complications is that it may involve persistent virus or viral
antigens and RNA in tissues, leading to chronic inammation. This can
trigger autoimmunity following an acute viral infection, as well as
dysbiosis of the microbiome and irreversible tissue damage. All of these
factors, to some degree or another, involve the immune system [31].
Previous studies indicate elevated PD-L1 levels in COVID-19 patients
with severe symptoms [32]. However, studies on soluble PD-L1 are
limited to L. Chavez-Galan et al demonstrated that increased sPD-L1
levels in COVID-19 patients are associated with requiring mechanical
ventilation [33]. F. Sabbatino et al showed that higher levels of sPD-L1
were found in deceased COVID-19 patients compared to survivors [16].
Our study suggests that the levels of sPD-L1 continue to be linked with
mortality even after one year of hospitalization.
The immune system is damaged during COVID-19 due to a failed
antiviral activity of interferons, which results in systemic manifesta-
tions. Additionally, there is higher degranulation of neutrophils, and
increased levels of cytokines, both in acute and long-term periods
[3437]. Combining different biomarkers shows promise in assessing
COVID-19 patient outcomes. During the rst wave of the COVID-19
pandemic, lymphopenia and NLR emerged as early prognostic markers
[38,39]. The biomarker NLR is a crucial component of the risk score
used to predict the likelihood of developing critical illness from COVID-
19 [38,40,41]. Its presence is explained by its contribution to the
pathogenesis of inammation and its related complications in a broader
sense.
Our study showed that the combination of two immune-mediated
markers (sPD-L1 and NLR), which reect the systemic inammatory
balance of activation and exhaustion, can complement each other and
improve the assessment of the risk of death in patients with COVID-19.
With a plasma sPD-L1 value of more than 277.13 pg/ml, the risk of
mortality within one year HR was 2.46, and for NLR more than 2.46 HR
was 2.87. Combining sPD-L1 and NLR while creating a model resulted in
a signicant improvement in the predictive accuracy of the Hazard
Ratio, which was found to be 7.6 (95 % CI: 3.0219.11).
It can be assumed that patients with more severe disease are at
higher risk of death, and therefore there is overlap in groups depending
Table 2
ROC-test characteristics. Signicance of sPD-L1 and NLR at admission in predicting mortality in patients with COVID-19.
Cut-off Youdens J AUC (95 % CI) Sensitivity, % (95 % CI) Specicity, % (95 % CI) p-value
sPD-L1 >277.13 0.2395 0.609 (0.5520.663) 57.89 (40.8 73.7) 66.06 (60.1 71.6) 0.033
NLR >2.46 0.2737 0.684 (0.6420.723) 77.7 (60.8 89.9) 49.59 (45.1 54.1) 0.0001
Fig. 3. Survival analysis of patients with COVID-19 for high versus low con-
centrations of (A) NLR, (B) sPD-L1, (C) integration of sPD-L1 and NLR. Hazard
ratios were calculated with the log-rank (Mantel-Cox) test.
Table 3
Multivariate Regression Model for Mortality in patients with COVID-19.
Variable OR (95 % Condence Interval) p-value
Sex (Male) 1.334 (0.6002.965) 0.480
Age over 60 years 4.183 (1.51311.599) 0.006
Charlson Comorbidity Index 2.489 (0.7258.542) 0.147
Severity illness 1.990 (0.8614.599) 0.108
High sPDL-1 and High NLR 2.359 (1.0255.429) 0.044
*OR =Odds ratio.
L. Akhmaltdinova et al.
International Immunopharmacology 129 (2024) 111600
6
on disease severity and mortality. It should be noted that sPD-L1 and
NLR concentrations (Fig. A1) in the non-survivor group did not differ
according to disease severity. It is suggested that elevated plasma con-
centrations of sPD-L1 and NLR are associated with increased mortality,
regardless of disease severity.
The importance of these ndings relates to the possibility of using
PDL-1 as a therapeutic target. Blockade of the PD-1/PDL-1 pathway is a
known therapeutic target in oncology, and
α
-PDL-1 antibody-based
therapies are currently in ongoing clinical trials to evaluate their ef-
fect on severe sepsis/septic shock [42]. Our study supports the theory
that specic anti-PDL therapy can contribute to both short-term and
long-term mortality reduction in COVID-19. Futrhermore, there is
another way to utilize this biomarker. The manifestations of COVID-19
vary widely and the severity of the disease is not always predictable.
Accordingly, personalizing therapy using PDL-1 inhibitors and cortico-
steroid therapy for patients with acute infections and post-infectious
therapy that induces PD-1/PDL-1 expression can improve patient
outcomes.
Our study found that age was not a signicant factor in disease
severity. However, we did observe a higher mean age in the group of
deceased patients, indicating that age may be a contributing factor to
poor outcomes (p =0.0001). Comorbidity was higher in both the group
of severe patients and the deceased. The differences in CRP, dimer, and
ferritin levels correspond with other studies based on disease severity
and outcome [43,44] A. Mainous et al. showed that increased levels of
CRP, which is one of the indicators of severe COVID-19 in the acute
period, are associated with an increased risk of mortality after 12
months of follow-up [45]. There were signicant differences in the main
inammatory markers CRP and D-dimer between survivors and
deceased, as well as between severe and moderate cases. Additionally,
ESR and ferritin levels also differed.
Inevitably, our study has several limitations. Firstly, we did not
investigate genetic variations in PD-L1 that may affect the expression
and function of this biomolecule [46,47]. Secondly, our study was
limited by the lack of analysis of how increased mortality from various
causes is associated with sPD-L1 and NLR levels. In this study, cardio-
vascular failure and acute vascular events were found to be the most
common causes of death in the post-COVID period. The limited number
of observations in each group was the main limitation to conducting a
detailed analysis of sPD-L1 and NLR concentrations based on mortality
causes. The study of all-cause mortality allowed for the inclusion of
those with conrmed COVID-19 pneumonia who died within one year.
Although another limitation such as examining only mortality as a single
endpoint, its signicant advantage is the relatively extended study
duration in the context of COVID-19 research.
5. Conclusions
Soluble PD-L1 and NLR represent distinct aspects of the inamma-
tory response and are involved in the dysregulated inammatory state
observed in COVID-19 patients. The study results suggest that a com-
bination of these biomarkers may be used to stratify patients with poor
outcome.
Ethics statement.
The study was conducted in accordance with the Declaration of
Helsinki, and approved by the Bioethics Committee of Karaganda
Medical University No. 18, dated 14 April 2021. Written informed
consent was obtained from all patients/participants who took part in
this study.
Funding.
This work was supported by the Ministry of Health of the Republic of
Kazakhstan, Program No. BR11065386.
CRediT authorship contribution statement
Lyudmila Akhmaltdinova: Writing original draft, Visualization,
Validation, Methodology, Formal analysis, Conceptualization. Irina
Mekhantseva: Writing original draft, Visualization, Validation, Soft-
ware, Methodology, Formal analysis, Data curation, Conceptualization.
Lyudmila Turgunova: Writing original draft, Visualization, Valida-
tion, Methodology, Formal analysis, Conceptualization. Mikhail Kos-
tinov: Writing review & editing, Conceptualization. Zhibek
Zhumadilova: Methodology, Investigation, Data curation. Anar Tur-
mukhambetova: Supervision, Resources, Project administration,
Funding acquisition.
Data availability
Data will be made available on request.
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.
org/10.1016/j.intimp.2024.111600.
References
[1] A.G. Mainous, B.J. Rooks, V. Wu, F.A. Orlando, COVID-19 post-acute sequelae
among adults: 12 month mortality risk, Front Med (lausanne). 8 (2021), https://
doi.org/10.3389/fmed.2021.778434.
[2] E.Y.F. Wan, S. Mathur, R. Zhang, V.K.C. Yan, F.T.T. Lai, C.S.L. Chui, X. Li, C.K.
H. Wong, E.W.Y. Chan, K.H. Yiu, I.C.K. Wong, Association of COVID-19 with short-
and long-term risk of cardiovascular disease and mortality: a prospective cohort in
UK Biobank, Cardiovasc Res. 119 (2023) 17181727, https://doi.org/10.1093/
cvr/cvac195.
[3] C. Qin, L. Zhou, Z. Hu, S. Zhang, S. Yang, Y. Tao, C. Xie, K. Ma, K. Shang, W. Wang,
D.-S. Tian, Dysregulation of Immune Response in Patients With Coronavirus 2019
(COVID-19) in Wuhan, China, Clin. Infect. Dis. 71 (2020) (2019) 762768, https://
doi.org/10.1093/cid/ciaa248.
[4] V. Di Noia, A. DAveni, M. Squadroni, G.D. Beretta, G.L. Ceresoli, Immune
checkpoint inhibitors in SARS-CoV-2 infected cancer patients: the spark that ignites
the re? Lung Cancer 145 (2020) 208210, https://doi.org/10.1016/j.
lungcan.2020.05.006.
[5] J. Vitte, A.B. Diallo, A. Boumaza, A. Lopez, M. Michel, J. Allardet-Servent,
S. Mezouar, Y. Sereme, J.-M. Busnel, T. Miloud, F. Malergue, P.-E. Morange,
P. Halfon, D. Olive, M. Leone, J.-L. Mege, A granulocytic signature identies
COVID-19 and its severity, J Infect Dis. 222 (2020) 19851996, https://doi.org/
10.1093/infdis/jiaa591.
[6] H.-L. Chang, P.-J. Wei, K.-L. Wu, H.-L. Huang, C.-J. Yang, Checkpoint inhibitor
pneumonitis mimicking COVID-19 infection during the COVID-19 pandemic, Lung
Cancer 146 (2020) 376377, https://doi.org/10.1016/j.lungcan.2020.06.013.
[7] E.V. Robilotti, N.E. Babady, P.A. Mead, T. Rolling, R. Perez-Johnston,
M. Bernardes, Y. Bogler, M. Caldararo, C.J. Figueroa, M.S. Glickman, A. Joanow,
A. Kaltsas, Y.J. Lee, A. Lucca, A. Mariano, S. Morjaria, T. Nawar, G.
A. Papanicolaou, J. Predmore, G. Redelman-Sidi, E. Schmidt, S.K. Seo,
K. Sepkowitz, M.K. Shah, J.D. Wolchok, T.M. Hohl, Y. Taur, M. Kamboj,
Determinants of COVID-19 disease severity in patients with cancer, Nat Med. 26
(2020) 12181223, https://doi.org/10.1038/s41591-020-0979-0.
[8] C.L. Day, D.E. Kaufmann, P. Kiepiela, J.A. Brown, E.S. Moodley, S. Reddy, E.
W. Mackey, J.D. Miller, A.J. Leslie, C. DePierres, Z. Mncube, J. Duraiswamy,
B. Zhu, Q. Eichbaum, M. Altfeld, E.J. Wherry, H.M. Coovadia, P.J.R. Goulder,
P. Klenerman, R. Ahmed, G.J. Freeman, B.D. Walker, PD-1 expression on HIV-
specic T cells is associated with T-cell exhaustion and disease progression, Nature
443 (2006) 350354, https://doi.org/10.1038/nature05115.
[9] C.E. Puronen, E.S. Ford, T.S. Uldrick, Immunotherapy in people with HIV and
Cancer, Front Immunol. 10 (2019), https://doi.org/10.3389/mmu.2019.02060.
[10] G. Sch¨
onrich, M.J. Raftery, The PD-1/PD-L1 Axis and Virus infections: a delicate
balance, Front Cell Infect Microbiol. 9 (2019), https://doi.org/10.3389/
fcimb.2019.00207.
[11] A. Dipasquale, P. Persico, E. Lorenzi, D. Rahal, A. Santoro, M. Simonelli, COVID-19
lung injury as a primer for immune checkpoint inhibitors (ICIs)-related pneumonia
in a patient affected by squamous head and neck carcinoma treated with PD-L1
blockade: a case report, J Immunother Cancer. 9 (2021) e001870.
[12] J. Chen, L. Vitetta, Increased PD-L1 expression may be associated with the cytokine
storm and CD8+T-cell exhaustion in severe COVID-19, J Infect Dis. 223 (2021)
16591660, https://doi.org/10.1093/infdis/jiab061.
[13] B. Diao, C. Wang, Y. Tan, X. Chen, Y. Liu, L. Ning, L. Chen, M. Li, Y. Liu, G. Wang,
Z. Yuan, Z. Feng, Y. Zhang, Y. Wu, Y. Chen, Reduction and functional exhaustion of
T cells in patients with coronavirus disease 2019 (COVID-19), Front Immunol. 11
(2020), https://doi.org/10.3389/mmu.2020.00827.
[14] S. Bellesi, E. Metafuni, S. Hohaus, E. Maiolo, F. Marchionni, S. DInnocenzo, M. La
Sorda, M. Ferraironi, F. Ramundo, M. Fantoni, R. Murri, A. Cingolani, S. Sica,
A. Gasbarrini, M. Sanguinetti, P. Chiusolo, V. De Stefano, Increased CD95 (Fas) and
PD-1 expression in peripheral blood T lymphocytes in COVID-19 patients, Br J
Haematol. 191 (2020) 207211, https://doi.org/10.1111/bjh.17034.
L. Akhmaltdinova et al.
International Immunopharmacology 129 (2024) 111600
7
[15] D.R. Beserra, R.W. Alberca, A.C.C.C. Branco, L. de Mendonça Oliveira, M.M. de
Souza Andrade, S.C. Gozzi-Silva, F.M.E. Teixeira, T.M. Yendo, A.J. da Silva Duarte,
M.N. Sato, Upregulation of PD-1 expression and high sPD-L1 levels associated with
COVID-19 severity, J Immunol Res. 2022 (2022) 19, https://doi.org/10.1155/
2022/9764002.
[16] F. Sabbatino, V. Conti, G. Franci, C. Sellitto, V. Manzo, P. Pagliano, E. De Bellis,
A. Masullo, F.A. Salzano, A. Caputo, I. Peluso, P. Zeppa, G. Scognamiglio, G. Greco,
C. Zannella, M. Ciccarelli, C. Cicala, C. Vecchione, A. Filippelli, S. Pepe, PD-L1
dysregulation in COVID-19 patients, Front Immunol. 12 (2021), https://doi.org/
10.3389/mmu.2021.695242.
[17] A.-L. Buicu, S. Cernea, I. Benedek, C.-F. Buicu, T. Benedek, Systemic inammation
and COVID-19 Mortality in patients with major noncommunicable diseases:
chronic coronary syndromes, Diabetes and Obesity, J Clin Med. 10 (2021) 1545,
https://doi.org/10.3390/jcm10081545.
[18] J.R. Ulloque-Badaracco, W. Ivan Salas-Tello, A. Al-kassab-C´
ordova, E.A. Alarc´
on-
Braga, V.A. Benites-Zapata, J.L. Magui˜
na, A.V. Hernandez, Prognostic value of
neutrophil-to-lymphocyte ratio in COVID-19 patients: a systematic review and
meta-analysis, Int J Clin Pract. 75 (2021), https://doi.org/10.1111/ijcp.14596.
[19] S. Sarkar, P. Khanna, A.K. Singh, The impact of neutrophil-lymphocyte count ratio
in COVID-19: a systematic review and meta-analysis, J Intensive Care Med. 37
(2022) 857869, https://doi.org/10.1177/08850666211045626.
[20] A. Karimi, P. Shobeiri, A. Kulasinghe, N. Rezaei, Novel systemic inammation
markers to predict COVID-19 prognosis, Front Immunol. 12 (2021), https://doi.
org/10.3389/mmu.2021.741061.
[21] Diagnosis and treatment protocol for COVID-19 patients (Trial Version 9), Health
Care Science. 1 (2022) 1428. 10.1002/hcs2.1.
[22] D. Radovanovic, B. Seifert, P. Urban, F.R. Eberli, H. Rickli, O. Bertel, M.A. Puhan,
P. Erne, Validity of Charlson Comorbidity Index in patients hospitalised with acute
coronary syndrome, in: Insights from the Nationwide AMIS plus Registry
20022012, 2014, pp. 288294, https://doi.org/10.1136/heartjnl-2013-304588.
[23] S. Bhaskar, A. Sinha, M. Banach, S. Mittoo, R. Weissert, J.S. Kass, S. Rajagopal, A.
R. Pai, S. Kutty, Cytokine storm in COVID-19immunopathological mechanisms,
clinical considerations, and therapeutic approaches: the REPROGRAM consortium
position paper, Front Immunol. 11 (2020), https://doi.org/10.3389/
mmu.2020.01648.
[24] J.S. Kim, J.Y. Lee, J.W. Yang, K.H. Lee, M. Effenberger, W. Szpirt, A. Kronbichler, J.
Il. Shin, Immunopathogenesis and treatment of cytokine storm in COVID-19,
Theranostics. 11 (2021) 316329, https://doi.org/10.7150/thno.49713.
[25] X.R. Shen, R. Geng, Q. Li, Y. Chen, S.F. Li, Q. Wang, J. Min, Y. Yang, B. Li, R.
D. Jiang, X. Wang, X.-S. Zheng, Y. Zhu, J.K. Jia, X.L. Yang, M.Q. Liu, Q.C. Gong, Y.
L. Zhang, Z.Q. Guan, H.L. Li, Z.H. Zheng, Z.L. Shi, H.L. Zhang, K. Peng, P. Zhou,
ACE2-independent infection of T lymphocytes by SARS-CoV-2, Signal Transduct
Target Ther. 7 (2022) 83, https://doi.org/10.1038/s41392-022-00919-x.
[26] S. Li, S. Li, C. Disoma, R. Zheng, M. Zhou, A. Razzaq, P. Liu, Y. Zhou, Z. Dong,
A. Du, J. Peng, L. Hu, J. Huang, P. Feng, T. Jiang, Z. Xia, SARS-CoV-2: Mechanism
of infection and emerging technologies for future prospects, Rev Med Virol. 31
(2021), https://doi.org/10.1002/rmv.2168.
[27] R.J. Jose, A. Manuel, COVID-19 cytokine storm: the interplay between
inammation and coagulation, Lancet, Respir Med. 8 (2020) e46e47, https://doi.
org/10.1016/S2213-2600(20)30216-2.
[28] J. Liu, S. Li, J. Liu, B. Liang, X. Wang, H. Wang, W. Li, Q. Tong, J. Yi, L. Zhao,
L. Xiong, C. Guo, J. Tian, J. Luo, J. Yao, R. Pang, H. Shen, C. Peng, T. Liu, Q. Zhang,
J. Wu, L. Xu, S. Lu, B. Wang, Z. Weng, C. Han, H. Zhu, R. Zhou, H. Zhou, X. Chen,
P. Ye, B. Zhu, L. Wang, W. Zhou, S. He, Y. He, S. Jie, P. Wei, J. Zhang, Y. Lu,
W. Wang, L. Zhang, L. Li, F. Zhou, J. Wang, U. Dittmer, M. Lu, Y. Hu, D. Yang,
X. Zheng, Longitudinal characteristics of lymphocyte responses and cytokine
proles in the peripheral blood of SARS-CoV-2 infected patients, EBioMedicine. 55
(2020) 102763, https://doi.org/10.1016/j.ebiom.2020.102763.
[29] S.H. Hamidi, S. Kadamboor Veethil, S.H. Hamidi, Role of pirfenidone in TGF-β
pathways and other inammatory pathways in acute respiratory syndrome
coronavirus 2 (SARS-CoV-2) infection: a theoretical perspective, Pharmacol. Rep.
73 (2021) 712727, https://doi.org/10.1007/s43440-021-00255-x.
[30] V.J. Costela-Ruiz, R. Illescas-Montes, J.M. Puerta-Puerta, C. Ruiz, L. Melguizo-
Rodríguez, SARS-CoV-2 infection: the role of cytokines in COVID-19 disease,
Cytokine Growth Factor Rev. 54 (2020) 6275, https://doi.org/10.1016/j.
cytogfr.2020.06.001.
[31] M. Merad, C.A. Blish, F. Sallusto, A. Iwasaki, The immunology and
immunopathology of COVID-19, Sci. 375 (2022) (1979) 11221127, https://doi.
org/10.1126/science.abm8108.
[32] S.R. Bonam, H. Hu, J. Bayry, Role of the PD-1 and PD-L1 axis in COVID-19, Future
Microbiol. 17 (2022) 985988, https://doi.org/10.2217/fmb-2022-0103.
[33] L. Chavez-Galan, A. Ruiz, K. Martinez-Espinosa, H. Aguilar-Duran, M. Torres,
R. Falfan-Valencia, G. P´
erez-Rubio, M. Selman, I. Buendia-Roldan, Circulating
levels of PD-L1, TIM-3 and MMP-7 Are promising biomarkers to differentiate
COVID-19 patients that require invasive mechanical ventilation, Biomolecules. 12
(2022) 445, https://doi.org/10.3390/biom12030445.
[34] A.C. Borczuk, R.K. Yantiss, The pathogenesis of coronavirus-19 disease, J Biomed
Sci. 29 (2022) 87, https://doi.org/10.1186/s12929-022-00872-5.
[35] S. Cambier, M. Metzemaekers, A.C. de Carvalho, A. Nooyens, C. Jacobs,
L. Vanderbeke, B. Malengier-Devlies, M. Gouwy, E. Heylen, P. Meersseman,
G. Hermans, E. Wauters, A. Wilmer, D. Schols, P. Matthys, G. Opdenakker, R.
E. Marques, J. Wauters, J. Vandooren, P. Proost, Atypical response to bacterial
coinfection and persistent neutrophilic bronchoalveolar inammation distinguish
critical COVID-19 from inuenza, JCI Insight. 7 (2022), https://doi.org/10.1172/
jci.insight.155055.
[36] D. McGonagle, K. Sharif, A. ORegan, C. Bridgewood, The role of cytokines
including interleukin-6 in COVID-19 induced pneumonia and macrophage
activation syndrome-like disease, Autoimmun Rev. 19 (2020) 102537, https://doi.
org/10.1016/j.autrev.2020.102537.
[37] S. Li, L. Jiang, X. Li, F. Lin, Y. Wang, B. Li, T. Jiang, W. An, S. Liu, H. Liu, P. Xu,
L. Zhao, L. Zhang, J. Mu, H. Wang, J. Kang, Y. Li, L. Huang, C. Zhu, S. Zhao, J. Lu,
J. Ji, J. Zhao, Clinical and pathological investigation of patients with severe
COVID-19, JCI Insight. 5 (2020), https://doi.org/10.1172/jci.insight.138070.
[38] A.-P. Yang, J. Liu, W. Tao, H. Li, The diagnostic and predictive role of NLR, d-NLR
and PLR in COVID-19 patients, Int Immunopharmacol. 84 (2020) 106504, https://
doi.org/10.1016/j.intimp.2020.106504.
[39] G. Ponti, M. Maccaferri, C. Ruini, A. Tomasi, T. Ozben, Biomarkers associated with
COVID-19 disease progression, Crit Rev Clin Lab Sci. 57 (2020) 389399, https://
doi.org/10.1080/10408363.2020.1770685.
[40] M. Seyit, E. Avci, R. Nar, H. Senol, A. Yilmaz, M. Ozen, A. Oskay, H. Aybek,
Neutrophil to lymphocyte ratio, lymphocyte to monocyte ratio and platelet to
lymphocyte ratio to predict the severity of COVID-19, Am J Emerg Med. 40 (2021)
110114, https://doi.org/10.1016/j.ajem.2020.11.058.
[41] M.S. Asghar, M. Akram, F. Yasmin, H. Najeeb, U. Naeem, M. Gaddam, M.S. Jafri,
M.J. Tahir, I. Yasin, H. Mahmood, Q. Mehmood, R.R. Marzo, Comparative analysis
of neutrophil to lymphocyte ratio and derived neutrophil to lymphocyte ratio with
respect to outcomes of in-hospital coronavirus disease 2019 patients: a
retrospective study, Front Med (lausanne). 9 (2022), https://doi.org/10.3389/
fmed.2022.951556.
[42] R. Chen, L. Zhou, PD-1 signaling pathway in sepsis: does it have a future? Clin.
Immunol. 229 (2021) 108742 https://doi.org/10.1016/j.clim.2021.108742.
[43] I. Doykov J. H¨
allqvist K.C. Gilmour L. Grandjean K. Mills W.E. Heywood ‘The long
tail of Covid-19 - The detection of a prolonged inammatory response after a
SARS-CoV-2 infection in asymptomatic and mildly affected patients F1000Res. 9
(2020) 1349. 10.12688/f1000research.27287.1.
[44] J. Zhang, Y. Cao, G. Tan, X. Dong, B. Wang, J. Lin, Y. Yan, G. Liu, M. Akdis, C.
A. Akdis, Y. Gao, Clinical, radiological, and laboratory characteristics and risk
factors for severity and mortality of 289 hospitalized COVID-19 patients, Allergy.
76 (2021) 533550, https://doi.org/10.1111/all.14496.
[45] A.G. Mainous, B.J. Rooks, F.A. Orlando, The impact of initial COVID-19 episode
inammation among adults on mortality within 12 months post-hospital discharge,
Front Med (lausanne). 9 (2022), https://doi.org/10.3389/fmed.2022.891375.
[46] T. Zhang, H. Liu, L. Jiao, Z. Zhang, J. He, L. Li, L. Qiu, Z. Qian, S. Zhou, W. Gong,
B. Meng, X. Ren, H. Zhang, X. Wang, Genetic characteristics involving the PD-1/
PD-L1/L2 and CD73/A2aR axes and the immunosuppressive microenvironment in
DLBCL, J Immunother Cancer. 10 (2022) e004114.
[47] K. Georgiou, L. Chen, M. Berglund, W. Ren, N.F.C.C. de Miranda, S. Lisboa,
M. Fangazio, S. Zhu, Y. Hou, K. Wu, W. Fang, X. Wang, B. Meng, L. Zhang, Y. Zeng,
G. Bhagat, M. Nordenskj¨
old, C. Sundstr¨
om, G. Enblad, R. Dalla-Favera, H. Zhang,
M.R. Teixeira, L. Pasqualucci, R. Peng, Q. Pan-Hammarstr¨
om, Genetic basis of PD-
L1 overexpression in diffuse large B-cell lymphomas, Blood 127 (2016)
30263034, https://doi.org/10.1182/blood-2015-12-686550.
L. Akhmaltdinova et al.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Severe acute respiratory syndrome-associated coronavirus-2 (SARS-CoV-2) is the causal agent of coronavirus disease-2019 (COVID-19), a systemic illness characterized by variably severe pulmonary symptoms, cardiac conduction abnormalities, diarrhea, and gastrointestinal bleeding, as well as neurologic deficits, renal insufficiency, myalgias, endocrine abnormalities, and other perturbations that reflect widespread microvascular injury and a pro-inflammatory state. The mechanisms underlying the various manifestations of viral infection are incompletely understood but most data suggest that severe COVID-19 results from virus-driven perturbations in the immune system and resultant tissue injury. Aberrant interferon-related responses lead to alterations in cytokine elaboration that deplete resident immune cells while simultaneously recruiting hyperactive macrophages and functionally altered neutrophils, thereby tipping the balance from adaptive immunity to innate immunity. Disproportionate activation of these macrophages and neutrophils further depletes normal activity of B-cells, T-cells, and natural killer (NK) cells. In addition, this pro-inflammatory state stimulates uncontrolled complement activation and development of neutrophil extracellular traps (NETS), both of which promote the coagulation cascade and induce a state of “thrombo-inflammation”. These perturbations have similar manifestations in multiple organ systems, which frequently show pathologic findings related to microvascular injury and thrombosis of large and small vessels. However, the pulmonary findings in patients with severe COVID-19 are generally more pronounced than those of other organs. Not only do they feature inflammatory thromboses and endothelial injury, but much of the parenchymal damage stems from failed maturation of alveolar pneumocytes, interactions between type 2 pneumocytes and non-resident macrophages, and a greater degree of NET formation. The purpose of this review is to discuss the pathogenesis underlying organ damage that can occur in patients with SARS-CoV-2 infection. Understanding these mechanisms of injury is important to development of future therapies for patients with COVID-19, many of which will likely target specific components of the immune system, particularly NET induction, pro-inflammatory cytokines, and subpopulations of immune cells.
Article
Full-text available
COVID-19 has several mechanisms that can lead to lymphocyte depletion/exhaustion. The checkpoint inhibitor molecule programmed death protein 1 (PD-1) and its programmed death-ligand 1 (PDL-1) play an important role in inhibiting cellular activity as well as the depletion of these cells. In this study, we evaluated PD-1 expression in TCD4+, TCD8+, and CD19+ lymphocytes from SARS-CoV-2-infected patients. A decreased frequency of total lymphocytes and an increased PD-1 expression in TCD4+ and CD19+ lymphocytes were verified in severe/critical COVID-19 patients. In addition, we found a decreased frequency of total monocytes with an increased PD-1 expression on CD14+ monocytes in severe/critical patients in association with the time of infection. Moreover, we observed an increase in sPD-L1 circulant levels associated with the severity of the disease. Overall, these data indicate an important role of the PD-1/PDL-1 axis in COVID-19 and may provide a severity-associated biomarker and therapeutic target during SARS-CoV-2 infection.
Article
Full-text available
Severe COVID-19 patients display dysregulated expression of checkpoint molecules PD-1 and its ligand PD-L1, suggesting that these checkpoint molecules could be considered as prognostic markers and therapeutic targets in severe cases of COVID-19.
Article
Full-text available
Introduction and objectives: In patients with coronavirus disease 2019 (COVID-19), several abnormal hematological biomarkers have been reported. The current study aimed to find out the association of neutrophil to lymphocyte ratio (NLR) and derived NLR (dNLR) with COVID-19. The objective was to compare the accuracy of both of these markers in predicting the severity of the disease. Materials and methods: The study was conducted in a single-center having patients with COVID-19 with a considerable hospital stay. NLR is easily calculated by dividing the absolute neutrophil count (ANC) with the absolute lymphocyte count (ALC) {ANC/ALC}, while dNLR is calculated by ANC divided by total leukocyte count minus ANC {ANC/(WBC-ANC)}. Medians and interquartile ranges (IQR) were represented by box plots. Multivariable logistic regression was performed obtaining an odds ratio (OR), 95% CI, and further adjusted to discover the independent predictors and risk factors associated with elevated NLR and dNLR. Results: A total of 1,000 patients with COVID-19 were included. The baseline NLR and dNLR were 5.00 (2.91-10.46) and 4.00 (2.33-6.14), respectively. A cut-off value of 4.23 for NLR and 2.63 for dNLR were set by receiver operating characteristic (ROC) analysis. Significant associations of NLR were obtained by binary logistic regression for dependent outcome variables as ICU stay (p < 0.001), death (p < 0.001), and invasive ventilation (p < 0.001) while that of dNLR with ICU stay (p = 0.002), death (p < 0.001), and invasive ventilation (p = 0.002) on multivariate analysis when adjusted for age, gender, and a wave of pandemics. Moreover, the indices were found correlating with other inflammatory markers such as C-reactive protein (CRP), D-dimer, and procalcitonin (PCT). Conclusion: Both markers are equally reliable and sensitive for predicting in-hospital outcomes of patients with COVID-19. Early detection and predictive analysis of these markers can allow physicians to risk assessment and prompt management of these patients.
Article
Full-text available
Background Inflammation in the initial COVID-19 episode may be associated with post-recovery mortality. The goal of this study was to determine the relationship between systemic inflammation in COVID-19 hospitalized adults and mortality after recovery from COVID-19. Methods An analysis of electronic health records (EHR) for patients from 1 January, 2020 through 31 December, 2021 was performed for a cohort of COVID-19 positive hospitalized adult patients. 1,207 patients were followed for 12 months post COVID-19 episode at one health system. 12-month risk of mortality associated with inflammation, C-reactive protein (CRP), was assessed in Cox regressions adjusted for age, sex, race and comorbidities. Analyses evaluated whether steroids prescribed upon discharge were associated with later mortality. Results Elevated CRP was associated other indicators of severity of the COVID-19 hospitalization including, supplemental oxygen and intravenous dexamethasone. Elevated CRP was associated with an increased mortality risk after recovery from COVID-19. This effect was present for both unadjusted (HR = 1.60; 95% CI 1.18, 2.17) and adjusted analyses (HR = 1.61; 95% CI 1.19, 2.20) when CRP was split into high and low groups at the median. Oral steroid prescriptions at discharge were found to be associated with a lower risk of death post-discharge (adjusted HR = 0.49; 95% CI 0.33, 0.74). Discussion Hyperinflammation present with severe COVID-19 is associated with an increased mortality risk after hospital discharge. Although suggestive, treatment with anti-inflammatory medications like steroids upon hospital discharge is associated with a decreased post-acute COVID-19 mortality risk.
Article
Full-text available
Background Targeting the PD-1/PD-L1/L2 (programmed cell death protein 1/programmed cell death ligand 1/ligand 2) pathway combined with other immunosuppressive signalings, such as CD73/A2aR (A2a adenosine receptor) adenosine signaling, has emerged as a promising strategy for cancer treatment. The genetic characteristics of these immune checkpoints need to be further investigated in diffuse large B-cell lymphoma (DLBCL). Methods We performed whole-exome sequencing/targeted deep sequencing to investigate the genetic characteristics of PD-1/PD-L1/L2 and CD73/A2aR. The immunosuppressive effect of these two pathways on the tumor microenvironment was evaluated via RNA sequencing. Single-cell RNA sequencing was further applied to investigate the dysfunctional CD8+ T cells. In addition, multiplex immunofluorescence staining was used to quantitatively assess the expression of dysfunctional CD8 ⁺ T cells in DLBCL. Results SP140 was identified as a novel translocation partner for PD-L1, and a new inversion was detected between PD-L1 and PD-L2, both leading to the upregulation of PD-L1 expression. CD73 genetic mutations did not increase mRNA and protein expression. Patients with genetically altered CD73 tended to have a better overall survival than patients with wild-type CD73. Both PD-1/PD-L1 and CD73/A2aR signaling mediated the immunosuppressive microenvironment in DLBCL. The numbers of CD8 ⁺ T cells with PD-1 and A2aR expression were positively correlated with the number of dysfunctional CD8 ⁺ T cells (R ² =0.974, p = 0.013). According to the grades of dysfunctional CD8 ⁺ T cells we defined, grade 1 dysfunctional CD8 ⁺ T cells, with either PD-1 ⁺ or A2aR ⁺ , were significantly associated with poorer survival than grade 0 dysfunctional CD8 ⁺ T cells, with both PD-1 ⁻ and A2aR ⁻ ; and patients with grade 2 dysfunctional CD8 ⁺ T cells showed the worst clinical outcomes. Conclusions This study describes the additional genetic basis of PD-L1 overexpression and characterizes certain genetic alterations of CD73/A2aR in DLBCL. The degree of T-cell dysfunction is correlated with clinical outcomes. Strategies that reverse T-cell dysfunction by inhibiting PD-1/PD-L1/L2, particularly in combination with CD73/A2aR, may show potential as effective therapeutic options for DLBCL.
Article
Full-text available
Background: COVID-19 is an infectious disease caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Many COVID-19 patients require invasive mechanical ventilation (IMV) while others, even with acute respiratory failure, do not (NIMV). Therefore, we aimed to evaluate serum levels of MMP-7 and molecules related to exhausted T-cells as potential biomarkers to differentiate between IMV and NIMV patients. Methods: 105 patients diagnosed with COVID-19 and confirmed by RT-PCR for SARS-CoV-2 were divided into two groups according to the requirement for IMV. Serum levels of sPD-L1, sPD-L2, sTIM-3, sGal-9 and sMMP-7 were quantified by ELISA and correlated with clinical data. Twelve patients were followed up after eight months to compare the levels of the biomarkers between acute disease and post-COVID-19. Results: IMV patients experienced a lower PaO2/FiO2 (p < 0.0001) and a longer hospital stay (p < 0.0001), and exhibited higher levels of sPD-L1 (p < 0.05), sTIM-3 (p < 0.01) and sMMP-7 (p < 0.0001) when compared with NIMV patients. According to a ROC analysis, sMMP-7 had the highest sensitivity (78%) and specificity (76%) with a cut point of 4.5 ng/mL, followed by sTIM-3 and sPD-L1. Eight months post-COVID-19, IMV patients displayed a significant decrease in the initially high levels of sPD-L1, sTIM-3 and sGal-9, while sPD-L2 was increased, and sMMP-7 was unchanged. Conclusion: Circulating levels of sPD-L1, sTIM-3 and sMMP-7 are potential biomarkers of disease severity to distinguish patients requiring IMV. MMP-7 could also be a marker for the persistence of lung lesions post-COVID-19.
Article
Aims: This study aims to evaluate the short- and long-term associations between COVID-19 and development of cardiovascular disease (CVD) outcomes and mortality in the general population. Methods and results: A prospective cohort of patients with COVID-19 infection between 16 March 2020 and 30 November 2020 was identified from UK Biobank, and followed for up to 18 months, until 31 August 2021. Based on age (within 5 years) and sex, each case was randomly matched with up to 10 participants without COVID-19 infection from two cohorts-a contemporary cohort between 16 March 2020 and 30 November 2020 and a historical cohort between 16 March 2018 and 30 November 2018. The characteristics between groups were further adjusted with propensity score-based marginal mean weighting through stratification. To determine the association of COVID-19 with CVD and mortality within 21 days of diagnosis (acute phase) and after this period (post-acute phase), Cox regression was employed. In the acute phase, patients with COVID-19 (n = 7584) were associated with a significantly higher short-term risk of CVD {hazard ratio (HR): 4.3 [95% confidence interval (CI): 2.6- 6.9]; HR: 5.0 (95% CI: 3.0-8.1)} and all-cause mortality [HR: 81.1 (95% CI: 58.5-112.4); HR: 67.5 (95% CI: 49.9-91.1)] than the contemporary (n = 75 790) and historical controls (n = 75 774), respectively. Regarding the post-acute phase, patients with COVID-19 (n = 7139) persisted with a significantly higher risk of CVD in the long-term [HR: 1.4 (95% CI: 1.2-1.8); HR: 1.3 (95% CI: 1.1- 1.6)] and all-cause mortality [HR: 5.0 (95% CI: 4.3-5.8); HR: 4.5 (95% CI: 3.9-5.2) compared to the contemporary (n = 71 296) and historical controls (n = 71 314), respectively. Conclusions: COVID-19 infection, including long-COVID, is associated with increased short- and long-term risks of CVD and mortality. Ongoing monitoring of signs and symptoms of developing these cardiovascular complications post diagnosis and up till at least a year post recovery may benefit infected patients, especially those with severe disease.
Article
To further improve the diagnosis and treatment of COVID‐19, China National Health Commission and the National Administration of Traditional Chinese Medicine convened a group of experts to revise the relevant content of the Diagnosis and Treatment Protocol for COVID‐19 Patients (Trial Version 8) and developed the Diagnosis and Treatment Protocol for COVID‐19 Patients (Trial Version 9), summarizing the etiological characteristics, epidemiological characteristics, pathological changes, clinical features, diagnosis, clinical classification, population with high risk of severe/critical illnesses, early warning predictors for severe/critical illnesses, differential diagnosis, case identification and reporting, treatment, nursing, discharge criteria and precautions after discharge, patient transfer, control of nosocomial infection in medical institutions, and disease prevention.