Вы находитесь на странице: 1из 9

Platelet-to-White Blood Cell Ratio: A Prognostic Predictor for

90-Day Outcomes in Ischemic Stroke Patients with


Intravenous Thrombolysis

Zhibo Chen, MD,* Yuanyuan Huang, MD,* Shanshan Li, MD,† Jie Lin, MD,*
Wenyue Liu, MD,‡ Zhangna Ding, MD,* Xiang Li, MD,* Ying Chen, MD,§
Wanhui Pang, MD,* Dehao Yang, MD,* Zhongqian Su, MD,* Jia Li, MD,*
Yiyun Weng, MD,* and Xu Zhang, MD*

Background: This study is aimed to investigate the relationship between platelet-


to-white blood cell ratio (PWR) and 90-day outcomes in acute stroke patients with
intravenous thrombolysis (IVT). Materials and Methods: A retrospective analysis
was performed on 168 patients receiving IVT for acute ischemic stroke. Complete
blood count evaluation was conducted at admission before IVT. A modified Rankin
Scale (mRS) score of 3-6 at 90 days was considered an unfavorable outcome. Results:
A total of 168 patients were included from 2013 to 2015. The mean age of the sample
was 64.6 (±12.3) years, and 23.2% were women. The median baseline National In-
stitutes of Health Stroke Scale score was 7.5 (interquartile range [IQR] 8.0) and the
90-day mRS score was 2 (IQR 2). In our multivariate logistic regression model, a
PWR greater than 23.52 (odds ratio .454, 95% confidence interval: .212-.973, P < .050)
was a predictor of 90-day outcomes. In addition, there was a significant difference
in the PWR values of patients between favorable outcome and unfavorable outcome
in the large-artery atherosclerosis subtype (28.241 ± 11.581 and 21.899 ± 9.107, re-
spectively; P = .005). Conclusions: The PWR at admission predicts 90-day outcomes
in ischemic stroke patients with IVT. With the easy and routine use of hemogram
analysis, the PWR should be investigated in further prospective randomized con-
trolled trials of acute stroke. Key Words: Outcomes—ischemic stroke—intravenous
thrombolysis—platelet-to-white blood cell ratio.
© 2016 National Stroke Association. Published by Elsevier Inc. All rights reserved.

From the *Department of Neurology, The First Affiliated Hospi-


Introduction
tal of Wenzhou Medical University, Wenzhou, China; †Department
Stroke is the leading cause of mortality and morbid-
of Infection and Liver Diseases, Liver Research Center, The First Af-
filiated Hospital of Wenzhou Medical University, Wenzhou, China;
ity worldwide which contributes to the rising costs of
‡Department of Endocrinology, The First Affiliated Hospital of medical treatment and cure.1 Prompt treatment with re-
Wenzhou Medical University, Wenzhou, China; and §Department of combinant tissue plasminogen activator (rt-PA) by
Ultrasound, The First Affiliated Hospital of Wenzhou Medical Uni- intravenous thrombolysis (IVT) has been demonstrated
versity, Wenzhou 325000, China.
as the only existing early therapy to restore blood flow
Received April 2, 2016; revision received May 16, 2016; accepted
June 8, 2016.
before irreversible damage to brain cells has occurred and
Address correspondence to Yiyun Weng, MD & Xu Zhang, MD, to improve clinical recovery after acute ischemic stroke
Department of Neurology, The First Affiliated Hospital of Wenzhou (AIS) in some people.2,3 Therapeutic effects indicated by
Medical University, Wenzhou 325000, China. E-mail: drzhangxu@ functional outcomes differ among AIS patients who un-
126.com; wmudrzhangxu@163.com.
derwent treatment with IVT with rt-PA.
1052-3057/$ - see front matter
© 2016 National Stroke Association. Published by Elsevier Inc. All
Thromboembolism, the most common mechanism of
rights reserved. cerebrovascular occlusion, has been considered as a com-
http://dx.doi.org/10.1016/j.jstrokecerebrovasdis.2016.06.015 plication of blood vessel injury and systemic inflammatory

2430 Journal of Stroke and Cerebrovascular Diseases, Vol. 25, No. 10 (October), 2016: pp 2430–2438
PWR AS A PREDICTOR OF 90-DAY OUTCOME IN ISCHEMIC STROKE PATIENTS 2431
4
disorders. Platelets play a pivotal role in the process of Exclusion criteria were according to the protocol of the
thrombosis after they are activated under various con- Safe Implementation of Thrombolysis in Stroke-Monitoring
ditions, including atherosclerosis, inflammation, and Study,25 except for the 80-year-old age limit, history of
hemorheology changes.5,6 Activated platelets aggregate at stroke, and concomitant diabetes. Patients with an onset-
sites of damaged endothelial cells and participate in the to-treatment time (OTT) of 3-6 hours after symptom onset
development of atherosclerotic lesions, which ultimate- were also not excluded. 26 Exclusion criteria also
ly leads to intracranial bloodstream interruption by arterial included patients with hematologic disorders, immuno-
thrombosis arising from plaque rupture.7-9 Many inves- suppressant drug users, those with an infection history
tigators have elucidated that higher mean platelet volumes within 2 weeks before onset of stroke, a stroke history
(MPVs) imply severer stroke and worse prognosis,10-12 within 6 months, patients with a history of malignancy,
whereas lower platelet counts (PLTs) are presented in AIS incomplete follow-up, and a baseline modified Rankin
patients, and platelets are largely expended with the for- Scale (mRS) score higher than 2. One hundred sixty-
mation and development of thrombosis.13,14 eight patients were included and 22 patients excluded:
White blood cells (WBCs) are also implicated in the five were beyond the therapeutic window, nine had in-
pathophysiology of ischemic vascular disease.15 Leuko- complete follow-up, and eight had insufficient medical
cytes are also recruited with increased tissue damage in records.
the ischemia area and neutrophils release various inflam- Patients were treated following the guidelines of the
matory mediators, such as proteases and reactive oxygen Study Group for Neurologic Diseases of the Chinese
species, which result in endothelium damage and tissue Medical Association. Physical therapy and standardized
necrosis.16,17 Elevated peripheral total leukocyte and neu- rehabilitation were applied when the status of the pa-
trophil counts are also associated with stroke severity on tients were stable. The subtypes of stroke were classified
admission,18 a higher recurrence of ischemic stroke,19 and as large-artery atherosclerosis (LAA), cardioembolism (CE),
even poorer functional outcome.20 small-vessel disease (SVD), other determined etiology, and
Recent studies have also elaborated on the interac- undetermined etiology.27 Neuroimaging evaluation and
tions between platelets with leukocytes in ischemic neurofunctional assessments were taken by trained in-
stroke21 and acute coronary syndrome.22 This interplay vestigators who were blinded to patient distribution.
has been elucidated to be correlated with ischemia–
reperfusion injury.23,24 Early prediction for prognosis of
stroke patients with IVT is benefit to attracting the Clinical Characteristics and Functional Outcomes
attention of clinicians at the time of admission, which of the Patients
may influence the decision of treatment to improve Clinical characteristics of the patients included demo-
functional recovery. We for the first time analyzed graphic findings; medical history such as history of
peripheral platelet-to-white blood cell ratio (PWR) and previous stroke or transient ischemic attack (TIA), atrial
aimed to investigate whether this ratio on admission fibrillation, coronary artery disease (CAD), hyperten-
was associated with functional outcome for AIS with sion, diabetes mellitus, dyslipidemia, and atherosclerosis;
intravenous (IV) rt-PA and might be an independent smoking and drinking; medication before admission, which
risk factor for stroke with the other conventional risk included therapy with statins, antiplatelets, and antico-
factors. We also investigated the association between agulants; thrombolysis-related information including OTT,
the initial severity of acute stroke and this parameter in NIHSS score on admission, and incidence of symptom-
systematically documented patients. atic intracranial hemorrhage; baseline information including
blood pressure (BP) before treatments, baseline blood
Materials and Methods glucose, creatinine, baseline blood PLT, and WBC count;
lipid profile measured within 48 hours after admission,
Study Population
including total cholesterol, triglyceride, low-density li-
We conducted a single-center analysis reviewing the poprotein cholesterol (LDL-C), and high-density lipoprotein
medical records of consecutive AIS patients admitted to cholesterol (HDL-C); etiology of stroke; mRS score at 3
the Department of Neurology, The First Affiliated Hos- months; and dosage and type of statin administrated for
pital of Wenzhou Medical University, recruited from March at least 3 weeks after the stroke onset. Statin dosages in
2013 to December 2015, and received IVT after in- the present study were categorized into 2 levels accord-
formed consent. The diagnosis of AIS was confirmed by ing to the attainable reduction of LDL-C described in head-
computer tomography perfusion imaging or diffusion- to-head comparisons, because the efficiency of various
weighted magnetic resonance imaging; Intravenous rt- types of statin differs substantially.28 Atorvastatin 20 mg
PA (0.9 mg/kg up to a maximum of 90 mg) was used and rosuvastatin 10 mg were determined as low dose level;
with 10% of the total dosage as a bolus and the rest over atorvastatin 40 and 60 mg or rosuvastatin 20 mg were
1 h. No patient received antithrombotic agents within 24 determined as high dose level. All patients had a com-
hours after rt-PA infusion. puted tomography at admission and at 24 hours after
2432 Z. CHEN ET AL.
stroke onset, or sooner if neurological worsening was noted. 64.6 ± 12.3 with 23.2% being female. The median base-
Symptomatic intracranial hemorrhage was defined as any line NIHSS score was 7.5 (IQR 8.0) and the median 90-
hemorrhage with neurological deterioration, meaning an day mRS score was 2 (IQR 2). We established a PWR of
NIHSS score of 4 or higher than that at admission, or 23.52 or lower as a preliminary cutoff point for poor
hemorrhage leading to death.29 Our safety end point was outcome (mRS score 3-6) following AIS with IVT
unfavorable outcome (mRS score ≥ 3) and favorable (χ2 = 10.400; P = .001), and divided the patients into 2
outcome (mRS score < 2) at 3 months. groups: low PWR and high PWR. The characteristics and
laboratory findings in patients with low and high PWRs
Blood Sample Analysis are reported in Table 1. The NIHSS scores on admission
to the hospital of patients in the group with low PWR
A hemogram was evaluated using the peripheral venous
were significantly higher than those of patients in the group
blood samples taken on admission to the emergency de-
with high PWR (median 10.0 [IQR 8.5] versus median
partment. The blood sample was collected in a calcium
6.0 [IQR 7.0], P = .011). There were also significant dif-
ethylenediaminetetraacetic acid tube. Complete blood count
ferences in terms of mRS score (median 3.0 [IQR 2.0] versus
evaluation was performed in the hematology laboratory
median 2.0 [IQR 2.0], P = .003) and OTT (χ2 = 10.162,
of our hospital by the hemocounter (XE2100, Sysmex Co.,
P = .006). In addition, there were no statistical differ-
Kobe, Japan) to calculate the count of leukocytes and plate-
ences between the 2 groups in age, sex, hypertension,
lets. PWR values were calculated as the count ratio of
diabetes mellitus, CAD, smoking, drinking, hyperlipid-
platelets to leukocytes in peripheral blood. In addition,
emia, atrial fibrillation, previous stroke or TIA, stroke
other routine laboratory findings were observed in the
subtype, systolic BP, diastolic BP, blood glucose, serum
digital record systems in the hospital.
creatinine, total cholesterol, triglyceride, LDL-C, HDL-
C, prior drug use, maintenance of the dose level of statin,
Statistical Analysis incidence of hemorrhagic transformation, symptomatic ce-
Data were analyzed using SPSS software (version 17.0; rebral hemorrhage, mortality rate, and so on.
SPSS Inc., Chicago, IL) and presented as mean ± stan- After 3 months, 64 patients (38.1%) had a poor/death
dard deviation or median (interquartile range [IQR]). outcome (mRS score ≥ 3). The percentage of patients with
Distribution normality was analyzed with the Kolmogorov– unfavorable outcome was significantly different between
Smirnov test. The difference between the 2 groups was patients with low and high PWRs (50.6% versus 26.4%,
tested using independent Student t-tests for normally dis- P = .001; Table 1). In the univariate analysis for 90-day
tributed variables; the Mann–Whitney U-test was used unfavorable prognosis, atrial fibrillation, hyperlipid-
for the comparison of nonparametrically distributed vari- emia, NIHSS score on admission, OTT, stroke subtype,
ables. The difference between 3 groups was tested using dose level of statin use, hemorrhagic transformation, symp-
analysis of variance for normally distributed variables; tomatic cerebral hemorrhage, and PWR greater than 23.52
the Kruskal–Wallis test was used for the comparison of were selected into multivariate logistic regression because
nonparametrically distributed variables. The difference all had P values less than .10 (Table 2). As shown in
between categorical variables was determined using the Table 2, after adjusting the influences of the factors above
χ2 test. Mean with standard deviation, median with IQR, by multivariate logistic regression, PWR greater than 23.52
and percentages were used to describe the distribution was an independent predictor for unfavorable outcome
of continuous and categorical variables, respectively. The defined as an mRS score of 3 or higher at 3 months
receiver operating characteristic (ROC) curve was used (odds ratio .454, 95% confidence interval [CI]: .212-.973,
to evaluate the prognosis effect of PWR, PLT, and WBC P = .042).
count on clinical outcome for AIS patients with IVT and ROC curves of PLT, WBC, and PWR to prediction of
to determine the optimal cutoff value for PWR. Logistic 90-day outcome are shown in Figures 1-3. The values of
regression analysis was performed to determine predic- area under the curve were .595 (95% CI: .509-.681, P = .039)
tors of 90-day functional outcome, and variables with a for PLT, .615 (95% CI: .524-.706, P = .013) for WBC, and
probability value less than .10 in univariate logistic re- .652 (95% CI: .566-.739, P = .001) for PWR. The optimal
gression analysis were determined unbalanced clinical cutoff value for PWR, of which the value of the area under
characteristics. All the unbalanced clinical characteris- the curve is higher than that of either PLT or WBC, as
tics and established predictors were then analyzed using a predictor for 90-day unfavorable outcome was determined
multivariate logistic regression analysis. Statistical sig- as 23.52 in the ROC curve. On this level, the sensitivity
nificance was set at a P value less than .05. was 64.1% and the specificity was 61.5%. The positive
predictive value of a PWR of 23.52 or lower was 50.6%
and the negative predictive value was 73.6% (Fig 3). There
Results
was a statistically significant difference in 90-day out-
A total of 168 patients were reviewed and included in comes between cardioembolic and other groups (χ2 = 6.550,
this analysis. The mean age of the patient sample was P = .010; Table 3). However, the PWR values were not
PWR AS A PREDICTOR OF 90-DAY OUTCOME IN ISCHEMIC STROKE PATIENTS 2433
Table 1. Demographic characteristics and laboratory findings in patients with low and high PWRs

Characteristics Total (n = 168) Low PWR† (n = 81) High PWR‡ (n = 87) P value

Age (years), mean ± SD 64.6 ± 12.3 64.3 ± 12.7 64.8 ± 12.0 NS


Female, n (%) 39 (23.2) 19 (23.5) 20 (23.0) NS
Hypertension, n (%) 121 (72.0) 61 (75.3) 60 (69.0) NS
Diabetes mellitus, n (%) 50 (29.8) 25 (30.9) 25 (28.7) NS
CAD, n (%) 14 (8.3) 7 (8.6) 7 (8.0) NS
Smoking, n (%) 69 (41.1) 38 (46.9) 31 (35.6) NS
Drinking, n (%) 55 (32.7) 25 (30.9) 30 (34.5) NS
Hyperlipidemia, n (%) 113 (67.3) 60 (74.1) 53 (60.9) NS
Atrial fibrillation, n (%) 45 (26.8) 23 (28.4) 22 (25.3) NS
Prior stroke or TIA, n (%) 28 (16.7) 16 (19.8) 12 (13.8) NS
Baseline NIHSS score, median (IQR) 7.5 (8.0) 10.0 (8.5) 6.0 (7.0) .011
OTT .006
~180 min 46 (27.4) 13 (16.0) 33 (37.9)
181~270 min 89 (53.0) 49 (60.5) 40 (46.0)
271~360 min 33 (19.6) 19 (23.5) 14 (16.1)
Stroke subtype NS
Large-artery atherosclerosis 107 (63.7) 52 (64.2) 55 (63.2)
Small-vessel disease 13 (7.7) 3 (3.7) 10 (11.5)
Cardioembolic 45 (26.8) 24 (29.6) 21 (24.1)
Other or unknown cause 3 (1.8) 2 (2.5) 1 (1.1)
Systolic BP (mmHg), mean ± SD 153.9 ± 20.3 153.2 ± 20.9 154.5 ± 19.9 NS
Diastolic BP (mmHg), mean ± SD 84.1 ± 12.7 82.6 ± 12.8 85.5 ± 12.5 NS
Glucose (mmol/L), mean ± SD 7.92 ± 2.97 8.36 ± 3.30 7.51 ± 2.579 NS
Creatinine (μmol/L), mean ± SD 73.7 ± 20.9 75.6 ± 19.9 72.4 ± 21.9 NS
Total cholesterol (mmol/L), mean ± SD 5.151 ± 1.093 5.197 ± 1.033 5.108 ± 1.150 NS
Triglyceride (mmol/L), mean ± SD 1.563 ± .986 1.468 ± .838 1.652 ± 1.104 NS
HDL-C (mmol/L), mean ± SD 1.204 ± .334 1.236 ± .352 1.175 ± .316 NS
LDL-C (mmol/L), mean ± SD 3.182 ± .886 3.238 ± .856 3.129 ± .915 NS
Prior drugs, n (%)
Antiplatelet 35 (20.8) 19 (23.5) 16 (18.4) NS
Anticoagulant 6 (3.6) 2 (2.5) 4 (4.6) NS
Statin 6 (3.6) 4 (4.9) 2 (2.3) NS
Statin use, n (%) NS
Not use 3 (1.8) 2 (2.5) 1 (1.1)
Low level* 43 (25.6) 24 (29.6) 19 (21.8)
High level§ 122 (72.6) 55 (67.9) 67 (77.0)
Hemorrhagic transformation, n (%) 38 (22.6) 22 (27.2) 16 (18.4) NS
sICH, n (%) 5 (3.0) 4 (4.9) 1 (1.1) NS
Mortality rate, n (%) 6 (3.6) 5 (6.2) 1 (1.1) NS
mRS score, median (IQR) 2.0 (2.0) 3.0 (2.0) 2.0 (2.0) .003
Unfavorable outcome at 90 days, n (%) 64 (38.1) 41 (50.6) 23 (26.4) .001

Abbreviations: BP, blood pressure; CAD, coronary artery disease; HDL-C, high-density lipoprotein cholesterol; HT, hemorrhagic trans-
formation; IQR, interquartile range; LDL-C, low-density lipoprotein cholesterol; mRS, modified Rankin Scale; NIHSS, National Institutes
of Health Stroke Scale; NS, not significant; OTT, onset-to-treatment time; PWR, platelet-to-white blood cell ratio; SD, standard deviation;
sICH, symptomatic cerebral hemorrhage; TIA, transient ischemic attack.
*Low level: atorvastatin, 20 mg; or rosuvastatin, 10 mg.
†Low PWR: patients with PWR of 23.52 or lower.
‡High PWR: patients with PWR greater than 23.52.
§High level: atorvastatin, 40 and 60 mg; or rosuvastatin, 20 mg.

significantly different among LAA, small-vessel disease, were significantly higher than those of unfavorable outcome
and cardioembolic subtypes (26.107 ± 11.181 versus in LAA stroke (28.241 ± 11.581 versus 21.899 ± 9.107, P = .005;
29.347 ± 7.695 versus 24.505 ± 9.000, respectively; P = .323; Table 3) but were not significantly different in either the
Table 3). In addition, the PWR values of favorable outcome small-vessel disease or cardioembolic subtype.
2434 Z. CHEN ET AL.
Table 2. Logistic regression model with predictors of unfavorable outcome (n = 168)

Variables Unadjusted OR (95% CI) P value Adjusted OR (95% CI) P value

Age 1.022 (.996-1.049) .104 — —


Female 1.348 (.651-2.790) .421 — —
Hypertension .873 (.438-1.739) .698 — —
Diabetes mellitus .778 (.390-1.554) .477 — —
CAD .895 (.286-2.798) .848 — —
Smoking .874 (.463-1.650) .678 — —
Drinking 1.005 (.518-1.952) .987
Hyperlipidemia .565 (.293-1.091) .089
Atrial fibrillation 2.371 (1.182-4.759) .015
Prior stroke 1.269 (.557-2.891) .570 — —
NIHSS score on admission 1.294 (1.186-1.411) <.001 1.265 (1.157-1.383) <.001
OTT
~180 min (reference) — .015 — —
181~270 min .999 (.467-2.134) .998 — —
271~360 min 3.179 (1.253-8.070) .015 — —
Stroke subtype
Large-artery atherosclerosis (reference) — .035 — —
Small-vessel disease .359 (.075-1.705) .197 — —
Cardioembolic 2.254 (1.108-4.584) .025 — —
Other or unknown cause 3.944 (.346-44.972) .269 — —
Systolic BP 1.000 (.985-1.015) .976 — —
Diastolic BP 1.014 (.989-1.039) .284 — —
Glucose .997 (.897-1.107) .949 — —
Creatinine .999 (.984-1.014) .907 — —
Total cholesterol .908 (.681-1.212) .513 — —
Triglyceride .982 (.714-1.351) .911 — —
HDL-C 1.924 (.754-4.911) .171 — —
LDL-C .860 (.600-1.231) .409 — —
Prior drugs —
Antiplatelet .950 (.440-2.051) .896 —
Anticoagulant .806 (.143-4.534) .807 — —
Statin .806 (.143-4.534) .807 — —
Statin use dose level .271 (.138-.533) <.001 .373 (.167-.831) .016
Hemorrhagic transformation 2.499 (1.197-5.220) .015 — —
Symptomatic cerebral hemorrhage 6.867 (.750-62.865) .088 — —
PWR (>23.52)* .351 (.184-.669) .001 .454 (.212-.973) .042

Abbreviations: BP, blood pressure; CAD, coronary artery disease; CI, confidence interval; HDL-C, high-density cholesterol; LDL-C, low-
density cholesterol; min, minute; NIHSS, National Institutes of Health Stroke Scale; OR, odds ratio; OTT, onset-to-treatment time; PWR,
platelet-to-white blood cell ratio.
*PWR was included in the regression model as a dichotomous variable as above cutoff value (>23.52) and below or equal to the cutoff
value (≤23.52).

Discussion endogenous active phosphoric acid mediator intimately


related with arachidonic acid metabolism, is produced
Atherosclerosis has been the most common pathogen- by a variety of tissues and cells involved in allergy and
esis implicated in the occurrence of cerebral infarction. inflammatory reaction. With the activation and destruc-
At the site of damaged endotheliocytes, the exposure of tion of platelets, PAF is released into plasma and results
vascular collagen increases the contact area with plate- in further platelet aggregation. The ischemia of paren-
lets, contributing to the development of thrombosis. In chyma gives rise to excessive production of PAF, which
this process, the aggregation of platelets with fibrino- may be an endogenous toxic nerve agent.30,31 D’Erasmo
gen plays a crucial role.7,8 Among the parameters of et al32 has demonstrated that PLT decreased with in-
platelets, the increase of MPV reflects the degree of plate- creased MPV within 48 hours after onset of stroke and
let activation and the ability to promote the formation returned to normal level on the ninth day. PLTs in the
of blood clots.9,10 Platelet activating factor (PAF), an death group have also been illustrated by D’Erasmo et al33
PWR AS A PREDICTOR OF 90-DAY OUTCOME IN ISCHEMIC STROKE PATIENTS 2435

Figure 3. Receiver operating curve of platelet-to-white blood cell ratio


Figure 1. Receiver operating curve of platelet count to prediction of 90-
to prediction of 90-day outcome in acute ischemic stroke with intrave-
day outcome in acute ischemic stroke with intravenous thrombolysis. Associated
nous thrombolysis. Associated criterion: less than 23.52; sensitivity: 64.1%;
criterion: less than 203.50; sensitivity: 68.8%; specificity: 51.0%; positive
specificity: 61.5%; positive predictive value: 50.6%; negative predictive value:
predictive value: 46.3%; negative predictive value: 72.6%. Area under the
73.6%. Area under the curve: .652 (95% confidence interval .566-.739);
curve: .595 (95% confidence interval .509-.681); P = .039.
P = .001.

to be obviously lower than those in the surviving group,


which may be associated with the prognosis of isch- obvious correlation between PLT and long-term
emic stroke. However, subsequent research demonstrated prognosis.12,34 Recently, Mayda-Domac et al12 concen-
the definite decrease of PLT in ischemic stroke34 without trated on the pathogenicity and predictor of MPV for
cerebral infarction, and PLT has been demonstrated as
an independent risk factor for ischemic stroke, which was
different from previous studies.
Inflammation has been increasingly recognized as a key
contributor to the pathogenesis of atherosclerosis, throm-
bus formation, and ischemic stroke, in which various
elements of the immune system are intimately involved.4
Leukocytes, also called immune cells, play a significant
role in vascular inflammatory injury and secondary pa-
renchyma damage after the onset of stroke. Cytokines and
adhesion molecules regulate the increased migration of
leukocytes to the infarct area.15 Intraparenchymal peri-
vascular neutrophil migration, which occurs within 6-24
hours, is the first response to ischemic brain damage. A
variety of inflammatory mediators and toxic-effective sub-
stances released by immune cells, particularly neutrophils,
may exaggerate brain edema or directly promote the death
of neurons in ischemic and reperfused areas.17 In a clin-
ical investigation, early leukocytosis and neutrophilia have
been reported to be correlated with infarct volume as evalu-
ated by diffusion-weighted magnetic resonance imaging
among patients with AIS,35 and have also been associ-
Figure 2. Receiver operating curve of white blood cell count to predic-
ated with higher recurrences of stroke.19 Furthermore,
tion of 90-day outcome in acute ischemic stroke with intravenous thrombolysis.
Associated criterion: greater than 9.33; sensitivity: 48.4%; specificity: 77.9%;
Furlan et al20 suggested that high degrees of peripheral
positive predictive value: 57.4%; negative predictive value: 71.1%. Area WBC were a marker of poor neurological prognosis. Also,
under the curve: .615 (95% confidence interval .524-.706); P = .013. low lymphocyte counts with increased counts of monocytes
2436 Z. CHEN ET AL.
Table 3. A comparison of mean platelet-to-white blood cell ratio among stroke subtypes and the relationship between this ratio and
90-day functional outcome for each stroke subtype

Total Favorable outcome Unfavorable outcome


Stroke subtype Mean ± SD n (mean ± SD) n (mean ± SD) P value*

Large-artery atherosclerosis (n = 107) 26.107 ± 11.181 71 (28.241 ± 11.581) 36 (21.899 ± 9.107) .005†
Cardioembolic (n = 45) 24.505 ± 9.000 21 (26.314 ± 7.561) 24 (22.922 ± 9.981) .211
Small-vessel disease (n = 13) 29.347 ± 7.695 11 (29.224 ± 8.421) 2 (30.025 ± .295) .899
P value* .323 .701 .481 —
χ2 6.550 — — .010‡

Abbreviation: SD, standard deviation.


*Comparisons of mean platelet-to-white blood cell ratio of different outcomes of patients for each stroke subtype and different subtypes
of patients with favorable or unfavorable outcome 90-day clinical outcome.
†There was a statistically significant difference in the mean platelet-to-white blood cell ratio between favorable and unfavorable outcomes
for the large-artery atherosclerosis subtype (P = .005).
‡There was a statistically significant difference in 90-day outcomes between cardioembolic and other stroke subtypes (P = .010).

have been proposed by Klehmet et al36 to contribute to leukocytes.45 Activated platelets also release a variety of
increasing physical susceptibility to poststroke infec- prothrombotic and proinflammatory mediators associ-
tion, leading to prolonged length of stay and aggravated ated with leukocyte recruitment, including soluble CD40
pathogenetic condition. ligand, regulated upon activation, normal T cell ex-
Increased MPV and platelet distribution width have also pressed and secreted (RANTES), and thromboxane A2.21
been reported to be associated with thrombolysis failure Moreover, the formation of LPA may contribute to
in patients with ST-segment elevation myocardial ischemia–reperfusion injury,23,24 which may be associ-
infarction.37 Previous studies have demonstrated lower ated with the therapeutic effect of IVT. Kupatt et al46 have
PLTs and larger platelet volumes in patients with acute proposed that LPAs have the properties to exaggerate
coronary syndrome38 and an inverse relationship between reperfusion injury by accumulating in blood vessels with
PLTs and MPVs in normal populations.39,40 Larger plate- an ischemic event, and Ritter et al24 have found an ap-
lets, with higher physiological activity and greater proximately 2-fold increase in LPA after ischemic stroke
glycoprotein IIb-IIIa receptor expression, contain more and reperfusion compared to preischemic values, leading
prothrombotic mediators and release increased active sub- to microvascular plugging and decreased ischemia per-
stances, which have potent properties to promote fusion. In this process, leukocytes are recruited with platelet
thrombosis5-7 and may influence the recanalization after aggregation and consumption. Thus, the PWR may rep-
IVT. Also, elevated leukocytes are correlated with resent the degree of inflammatory and thrombotic events,
thromboresistance, reduced epicardial bloodstream, and and reflect the severity of ischemic stroke and progno-
myocardial perfusion, and adverse prognosis of acute myo- sis of patients treated with IVT.
cardial infarction after thrombolytic administration.41 In To our knowledge, this is the first study to calculate
experimental models of myocardial ischemia–reperfusion, the PWR and investigate the relationship between this
leukocytes may contribute to the endothelial dysfunc- ratio and 90-day outcomes for AIS with IVT. The results
tion and microvascular obstruction, leading to low of our retrospective study indicate that a higher WBC
myocardial perfusion status,42,43 which is also described with a lower PLT on admission is associated with poorer
in rat models of ischemic stroke and reperfusion.23,24 prognosis with respect to initial stroke severity (NIHSS
Platelet–leukocyte interactions have been increasingly score) on admission and 90-day functional outcome (mRS
elucidated to be associated with inflammation and onset score). The results were suitably adjusted for several major
of ischemic events.21,44 Leukocyte–platelet aggregates (LPAs) potential confounders, including age, sex, hypertension,
in peripheral blood have been considered as a novel marker diabetes mellitus, CAD, smoking, drinking, hyperlipid-
of activated platelets,15 which possess the “bridge” effects emia, atrial fibrillation, prior stroke or TIA, prior drug
for other cells in vasculature, particularly leukocytes re- use, maintenance of the dose level of statin, OTT, stroke
cruited via platelet secretory components including several subtype, hemorrhagic transformation, symptomatic ce-
chemokines and membrane ligands.21 The interplay of plate- rebral hemorrhage, baseline NIHSS score, BP, glucose,
lets and leukocytes is primarily attributed to P-selectin, creatinine, and blood lipid profiles. Moreover, our study
an adhesive molecule reserved in platelet alpha-granules provides the best estimated cutoff value of PWR for pre-
and transported to the surface of activated platelets. The diction of 90-day outcomes after AIS with thrombolytic
pivotal receptor for P-selectin, P-selectin glycoprotein ligand- therapy. Also, there was an evident difference in the PWR
1, is elementarily expressed on the membranes of most values between favorable and unfavorable outcomes in
PWR AS A PREDICTOR OF 90-DAY OUTCOME IN ISCHEMIC STROKE PATIENTS 2437
patients with the LAA subtype. The predictive effect of 4. Deb P, Sharma S, Hassan KM. Pathophysiologic
PWR on prognosis was independent of other risk factors mechanisms of acute ischemic stroke: an overview with
for ischemic stroke patients with IV rt-PA treatment, par- emphasis on therapeutic significance beyond thrombolysis.
Pathophysiology 2010;17:197-218.
ticularly LAA subtype. 5. Franco AT, Corken A, Ware J. Platelets at the interface
The present study has several limitations. The sever- of thrombosis, inflammation, and cancer. Blood
ity of stroke is indicated by NIHSS score47 and infarct 2015;126:582-588.
volume.30 We only found a correlation between PWR and 6. Walsh TG, Metharom P, Berndt MC. The functional role
NIHSS score, but an association between infarct volume of platelets in the regulation of angiogenesis. Platelets
2015;26:199-211.
and PWR could not be evaluated because no infarct volume 7. Alexandru N, Andrei E, Dragan E, et al. Interaction of
measurements were made. Also, the limited sample size platelets with endothelial progenitor cells in the
may be a limitation to the variable screening for multi- experimental atherosclerosis: role of transplanted
variate logistic regression and other statistical analyses, endothelial progenitor cells and platelet microparticles.
in particular the comparison of mean PWR values among Biol Cell 2015;107:189-204.
8. Fuentes QE, Fuentes QF, Andres V, et al. Role of platelets
stroke subtypes. It was determined that the 90-day out- as mediators that link inflammation and thrombosis in
comes of the CE subtype were poorer than those of other atherosclerosis. Platelets 2013;24:255-262.
subtypes, whereas the PWR values of the CE subtype were 9. Oz II, Yucel M, Bilici M, et al. Is mean platelet volume
not significantly lower than those of the LAA or SVD a reliable marker to predict ischemic stroke in the
subtype. Further, we observed the difference in PWR values follow-up of patients with carotid stenosis? J Stroke
Cerebrovasc Dis 2016;25:404-409.
between favorable and unfavorable outcomes for the LAA 10. Balcik OS, Bilen S, Ulusoy EK, et al. Thrombopoietin and
subtype. This trend, however, cannot be accurately evalu- mean platelet volume in patients with ischemic stroke.
ated in SVD and CE subtypes because of the sample- Clin Appl Thromb Hemost 2013;19:92-95.
size limitation. Moreover, some patients with prior 11. Ghahremanfard F, Asghari N, Ghorbani R, et al. The
hemogram analysis in other hospitals on admission, would relationship between mean platelet volume and severity
of acute ischemic brain stroke. Neurosciences (Riyadh)
not be drawn blood again and be excluded in the present 2013;18:147-151.
study, which may result in a selection bias for analysis. 12. Mayda-Domac F, Misirli H, Yilmaz M. Prognostic role
In conclusion, our study agrees with the clinical im- of mean platelet volume and platelet count in ischemic
portance of PLT and WBC count in ischemic stroke with and hemorrhagic stroke. J Stroke Cerebrovasc Dis
IVT and shows for the first time a clear correlation between 2010;19:66-72.
13. Jurk K, Jahn UR, Van Aken H, et al. Platelets in patients
PWR and initial stroke severity at the time of admis- with acute ischemic stroke are exhausted and refractory
sion. The association between PWR and outcomes for to thrombin, due to cleavage of the seven-transmembrane
ischemic stroke is independent of other known predic- thrombin receptor (PAR-1). Thromb Haemost 2004;91:334-
tive factors; thus, the study of PWR may enable patients 344.
to be identified at risk for future unfavorable prognosis. 14. Westrick R, Fredman G. Platelets: context-dependent
vascular protectors or mediators of disease. Arterioscler
This ratio, with an easy and routine measurement, may Thromb Vasc Biol 2015;35:e25-e29.
be an early, convenient, and important predictor for the 15. Ishikawa T, Shimizu M, Kohara S, et al. Appearance of
prognosis of ischemic stroke, particularly the LAA stroke WBC-platelet complex in acute ischemic stroke,
subtype. The value of PWR in AIS with or without IVT predominantly in atherothrombotic infarction. J Atheroscler
should be investigated in future prospective random- Thromb 2012;19:494-501.
16. Jickling GC, Liu D, Ander BP, et al. Targeting neutrophils
ized controlled trials. in ischemic stroke: translational insights from experimental
studies. J Cereb Blood Flow Metab 2015;35:888-901.
Acknowledgment: The present study was supported by the 17. Perez-de-Puig I, Miro-Mur F, Ferrer-Ferrer M, et al.
Foundation of Wenzhou Municipal Science and Technology Neutrophil recruitment to the brain in mouse and human
Bureau (No. Y20130214). ischemic stroke. Acta Neuropathol 2015;129:239-257.
18. Hug A, Dalpke A, Wieczorek N, et al. Infarct volume
is a major determiner of post-stroke immune cell function
and susceptibility to infection. Stroke 2009;40:3226-
References 3232.
19. Grau AJ, Boddy AW, Dukovic DA, et al. Leukocyte count
1. Mozaffarian D, Benjamin EJ, Go AS, et al. Executive as an independent predictor of recurrent ischemic events.
summary: heart disease and stroke statistics-2016 update: Stroke 2004;35:1147-1152.
a report from the American Heart Association. Circulation 20. Furlan JC, Vergouwen MD, Fang J, et al. White blood
2016;133:447-454. cell count is an independent predictor of outcomes after
2. Tissue plasminogen activator for acute ischemic stroke. acute ischaemic stroke. Eur J Neurol 2014;21:215-222.
The National Institute of Neurological Disorders and 21. Franks ZG, Campbell RA, Weyrich AS, et al. Platelet-
Stroke rt-PA Stroke Study Group. N Engl J Med leukocyte interactions link inflammatory and
1995;333:1581-1587. thromboembolic events in ischemic stroke. Ann N Y Acad
3. Wardlaw JM, Murray V, Berge E, et al. Thrombolysis for Sci 2010;1207:11-17.
acute ischaemic stroke. Cochrane Database Syst Rev 22. Szabo S, Etzel D, Ehlers R, et al. Combined thrombolysis
2014;(7):CD000213. with abciximab favourably influences platelet-leukocyte
2438 Z. CHEN ET AL.
interactions and platelet activation in acute myocardial 35. Buck BH, Liebeskind DS, Saver JL, et al. Early neutrophilia
infarction. J Thromb Thrombolysis 2005;20:155-161. is associated with volume of ischemic tissue in acute
23. Ritter LS, Orozco JA, Coull BM, et al. Leukocyte stroke. Stroke 2008;39:355-360.
accumulation and hemodynamic changes in the cerebral 36. Klehmet J, Harms H, Richter M, et al. Stroke-induced
microcirculation during early reperfusion after stroke. immunodepression and post-stroke infections: lessons from
Stroke 2000;31:1153-1161. the preventive antibacterial therapy in stroke trial.
24. Ritter LS, Stempel KM, Coull BM, et al. Leukocyte-platelet Neuroscience 2009;158:1184-1193.
aggregates in rat peripheral blood after ischemic stroke 37. Cetin M, Bakirci EM, Baysal E, et al. Increased platelet
and reperfusion. Biol Res Nurs 2005;6:281-288. distribution width is associated with ST-segment elevation
25. Wahlgren N, Ahmed N, Dávalos A, et al. Thrombolysis myocardial infarction and thrombolysis failure. Angiology
with alteplase for acute ischaemic stroke in the Safe 2014;65:737-743.
Implementation of Thrombolysis in Stroke-Monitoring 38. Ranjith MP, Divya R, Mehta VK, et al. Significance of
Study (SITS-MOST): an observational study. Lancet platelet volume indices and platelet count in ischaemic
2007;369:275-282. heart disease. J Clin Pathol 2009;62:830-833.
26. Ahmed N, Kellert L, Lees KR, et al. Results of intravenous 39. Bessman JD, Williams LJ, Gilmer PR Jr. Mean platelet
thrombolysis within 4.5 to 6 hours and updated results volume. The inverse relation of platelet size and count
within 3 to 4.5 hours of onset of acute ischemic stroke in normal subjects, and an artifact of other particles. Am
recorded in the Safe Implementation of Treatment in J Clin Pathol 1981;76:289-293.
Stroke International Stroke Thrombolysis Register 40. Lamparelli RD, Baynes RD, Atkinson P, et al. Platelet
(SITS-ISTR): an observational study. JAMA Neurol parameters. Part I. Platelet counts and mean platelet
2013;70:837-844. volume in normal and pregnant subjects. S Afr Med J
27. Adams HP Jr, Bendixen BH, Kappelle LJ, et al. 1988;73:36-39.
Classification of subtype of acute ischemic stroke. 41. Barron HV, Cannon CP, Murphy SA, et al. Association
Definitions for use in a multicenter clinical trial. TOAST. between white blood cell count, epicardial blood flow,
Trial of Org 10172 in Acute Stroke Treatment. Stroke myocardial perfusion, and clinical outcomes in the setting
1993;24:35-41. of acute myocardial infarction: a thrombolysis in
28. Scheitz JF, Seiffge DJ, Tutuncu S, et al. Dose-related effects myocardial infarction 10 substudy. Circulation
of statins on symptomatic intracerebral hemorrhage and 2000;102:2329-2334.
outcome after thrombolysis for ischemic stroke. Stroke 42. Engler RL, Schmid-Schonbein GW, Pavelec RS. Leukocyte
2014;45:509-514. capillary plugging in myocardial ischemia and reperfusion
29. Wardlaw JM, Murray V, Berge E, et al. Recombinant tissue in the dog. Am J Pathol 1983;111:98-111.
plasminogen activator for acute ischaemic stroke: an 43. Horwitz LD, Kaufman D, Kong Y. An antibody to
updated systematic review and meta-analysis. Lancet leukocyte integrins attenuates coronary vascular injury
2012;379:2364-2372. due to ischemia and reperfusion in dogs. Am J Physiol
30. Adunsky A, Hershkowitz M, Atar E, et al. Infarct volume, 1997;272:H618-H624.
neurological severity and PAF binding to platelets of 44. Marquardt L, Anders C, Buggle F, et al. Leukocyte-platelet
patients with acute cerebral ischemic stroke. Neurol Res aggregates in acute and subacute ischemic stroke.
1999;21:645-648. Cerebrovasc Dis 2009;28:276-282.
31. Belayev L, Khoutorova L, Atkins K, et al. LAU-0901, a 45. Huo Y, Xia L. P-selectin glycoprotein ligand-1 plays a
novel platelet-activating factor receptor antagonist, confers crucial role in the selective recruitment of leukocytes into
enduring neuroprotection in experimental focal cerebral the atherosclerotic arterial wall. Trends Cardiovasc Med
ischemia in the rat. Brain Res 2009;1253:184-190. 2009;19:140-145.
32. D’Erasmo E, Acca M, Celi FS, et al. Plasma fibrinogen 46. Kupatt C, Habazettl H, Hanusch P, et al. c7E3Fab reduces
and platelet count in stroke. J Med 1993;24:185-191. postischemic leukocyte-thrombocyte interaction mediated
33. D’Erasmo E, Aliberti G, Celi FS, et al. Platelet count, mean by fibrinogen. Implications for myocardial reperfusion
platelet volume and their relation to prognosis in cerebral injury. Arterioscler Thromb Vasc Biol 2000;20:2226-2232.
infarction. J Intern Med 1990;227:11-14. 47. Rost NS, Bottle A, Lee JM, et al. Stroke severity is a crucial
34. O’Malley T, Langhorne P, Elton RA, et al. Platelet size predictor of outcome: an international prospective
in stroke patients. Stroke 1995;26:995-999. validation study. J Am Heart Assoc 2016;5.

Вам также может понравиться