Next Article in Journal
Variability in Thyroid Cancer Multidisciplinary Team Meeting Recommendations Is Not Explained by Standard Variables: Outcomes of a Single Centre Review
Next Article in Special Issue
In Search of Clinical Impact: Advanced Monitoring Technologies in Daily Heart Failure Care
Previous Article in Journal
Biomarkers of Post-COVID Depression
Previous Article in Special Issue
Use of Extracorporeal Life Support for Heart Transplantation: Key Factors to Improve Outcome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Comparative Analysis of the Kinetic Behavior of Systemic Inflammatory Markers in Patients with Depressed versus Preserved Left Ventricular Function Undergoing Transcatheter Aortic Valve Implantation

Kaplan Heart Center, Kaplan Medical Center, Rehovot, Affiliated with the Hebrew University, Jerusalem 76610, Israel
*
Author to whom correspondence should be addressed.
These authors have contributed equally to this work.
J. Clin. Med. 2021, 10(18), 4148; https://doi.org/10.3390/jcm10184148
Submission received: 26 July 2021 / Revised: 20 August 2021 / Accepted: 5 September 2021 / Published: 15 September 2021
(This article belongs to the Special Issue Current Challenges and Advances in Heart Failure and Heart Transplant)

Abstract

:
Background: Prior studies have proven the safety and efficacy of transcatheter aortic valve implantation (TAVI) in patients with reduced left ventricular (LV) function. This study’s aim was to investigate periprocedural inflammatory responses after TAVI. Methods: Patients with severe symptomatic aortic stenosis and reduced LV function who underwent transfemoral TAVI were enrolled. A paired-matched analysis (1:2 ratio) was performed using patients with preserved LV function. Whole white blood cells (WBC) and subpopulation dynamics as well as the neutrophil to lymphocyte ratio (NLR) were evaluated at different times. Results: A total of 156 patients were enrolled, including 52 patients with LVEF < 40% 35.00 [30.00, 39.25] and 104 with LVEF > 50% 55.00 [53.75, 60.0], p < 0.001. Baseline NLR in the reduced LV function group was significantly higher compared to the preserved LV function group, 2.85 [2.07, 4.78] vs. 3.90 [2.67, 5.26], p < 0.04. After a six-month follow-up, the inflammatory profile was found to be similar in the two groups, NLR 2.94 [2.01, 388] vs. 3.30 [2.06, 5.35], p = 0.288. No significant mortality differences between the two groups were observed in the long-term outcome. Conclusions: TAVI for severe symptomatic aortic stenosis, with reduced LV function, was associated with an improvement in the inflammatory profile that may account for some of the observable benefits of the procedure in this subset of patients.

1. Introduction

Surgical aortic valve replacement performed in patients with reduced left ventricular ejection fraction (LVEF) is associated with worse patient outcomes [1,2]. However, transcatheter aortic valve implantation (TAVI) does not increase mortality or heart failure readmissions [3,4]. Moreover, patients with reduced LV function obtain better outcomes from TAVI compared to patients with preserved LV function [5,6,7,8].
Atherosclerosis and degenerative aortic stenosis have common risk factors and etiopathogenesis, and inflammation appears to play an important role in this respect. Furthermore, systemic inflammation has been recognized as a dominant feature in the progression of heart failure [9,10,11]. Specifically, inflammation has been linked to disease development, progression, and associated complications, and is predictive of poor patient outcomes, independent of LVEF [12,13,14]. One such inflammatory biomarker is the NLR, which is derived from routine complete blood counts. The NLR leverages early hematological observations in which the absolute neutrophil count is positively associated with cardiovascular events, while the absolute lymphocyte count is negatively associated with cardiovascular events [15,16]. Moreover, while total white blood cell counts have been shown to predict cardiovascular risk, the NLR appears to be a superior predictor [17]. Some studies have evaluated the NLR as a predictor of total mortality in the setting of acute coronary interventions [17,18] and heart failure [19].
We aimed to compare the kinetic behavior of inflammatory profile after TAVI in patients with preserved or reduced LV function by measuring the NLR biomarker as a potential mediator of outcome improvement from the procedure.

2. Methods

2.1. Study Population

We retrospectively included patients with severe symptomatic aortic stenosis who underwent TAVI in our center between January 2010 and March 2020. This study was approved by Kaplan Medical Center’s IRB. We excluded patients with chronic systemic inflammatory or autoimmune diseases, acute infections, hematological disorders, malignancies, dialysis, or treatment with agents affecting the white blood cell count, e.g., corticosteroids. Patients who did not have repeated blood tests and those with periprocedural death (up to 72 h after TAVI) were excluded as well.

2.2. TAVI Procedure

The type and size of the valves were at the discretion of the local heart team, and a decision was made according to the patient’s anatomical and clinical characteristics. Patients treated with BEV were implanted with either SAPIEN, SAPIEN XT, or S3 (Edwards Lifesciences, Irvine, CA, USA) valves. Patients treated with SEV were implanted with either CoreValve, Evolut PRO, or Evolut R (Medtronic, Inc., Minneapolis, MN, USA) valves. The small number of subjects treated with other valves were also excluded from the analysis. Transfemoral vascular access and closure was performed using the percutaneous approach with the safety wire technique and the Prostar XL (Abbott Vascular, Redwood City, CA, USA) vascular closure device. The procedure duration was calculated as “skin-to-skin”, i.e., time 0 marked the beginning of arterial blood pressure regulation by the accessory support access and the final time was represented by the termination of this accessory support access. Following that, we also used local anesthesia with conscious sedation as a first-line approach. All patients received unfractionated heparin to maintain a minimum active clotting time of >250 s after the insertion of the femoral sheet. Protamine (1 mg for each 100 U of heparin, maximal dose 50 mg) was administered at the time of vascular closure if needed. The use of prophylactic antibiotics during the procedure or hospital stay was routinely avoided. Aspirin was recommended before TAVI. Dual-antiplatelet treatment with 100 mg of aspirin and 75 mg of clopidogrel was started the day before the procedure and followed thereafter for six months, except for patients requiring chronic oral anticoagulation.

2.3. Inflammatory Markers

Baseline characteristics, procedural data, and clinical outcomes were collected. Blood samples were obtained using a 21G sterile syringe without stasis. Laboratory analyses were performed before the procedure, during the patient’s postprocedural intensive care unit stay on a daily basis, and at the physician’s discretion in the cardiology ward. These were retrospectively collected. The neutrophil–lymphocyte ratio (NLR) was calculated using this formula: NLR = absolute neutrophils count/absolute lymphocytes count.

2.4. Definition Criteria for Events

All the outcome definitions were strictly determined according to the Valve Academic Research Consortium 2 (VARC-2) criteria. All of these standardized endpoint definitions together represent the major adverse cardiovascular events (MACE) in our study. Clinical follow-up included 30-day and six-month visits after hospital discharge. The follow-ups were performed on site.

2.5. Matching

The reduced LV function cohort (LVEF < 40%) was compared to a preserved LV function cohort (LVEF > 50%), using matched-pair grouping for statistical analysis in a retrospective and descriptive manner. Patients suffering mid-range LV dysfunction, i.e., LVEF between 40% to 50%, or severely depressed LV function (LVEF ≤ 25%), were excluded. Matched patients were also selected during the same time period out of 104 patients undergoing TAVI. Age ± 5 years, sex, BMI ± 5, dialysis and additive EuroSCORE ± 5 were used for matched-pair analysis (1:2). Thus, a total of 156 patients were matched and compared. We performed a propensity score to build the matching groups.

2.6. Statistical Analysis

Data are presented as median values (with ranges in parentheses). Continuous variables between the various study groups were tested for normality with a Shapiro–Wilk test and when an abnormal distribution was found, a Mann–Whitney test was performed. When the distribution was normal, a t-test was used. Pearson’s chi-square test was performed for categorical variables when appropriate. Main effect estimates are presented with their 95% confidence interval. Statistical analysis was performed using SPSS software (IBM SPSS Statistics for Windows, Version 25.0., Armonk, NY, USA) and R programming version 4.0.2 (The R Foundation for Statistical Computing, Vienna, Austria; http://www.R-project.org) (accessed date 1 June 2021) for all analyses. Where the p values were less than 0.05, these were considered statistically significant.

3. Results

3.1. Pre-Procedural Data

During the ten-year study period, 370 consecutive patients were enrolled. The study flowchart is shown in Figure 1. According to our definitions, a total of 56 patients were excluded. The analyzed population included 314 patients (43% female, with a median age of 81.00 [76.00–85.00] years old), with severe symptomatic aortic stenosis (transaortic pressure gradient 37.00 [29.50–47.00] mmHg), and high or prohibitive operative risk (an STS score of 8.01 [5.1–10.3]). Baseline and procedural characteristics of the 1:2 matched study population according to ejection fraction are summarized in Table 1.
The clinical profile and executive procedural characteristics showed no significant differences between the two 1:2 matched population group types at baseline. As expected, according to the study aims, compared with patients with preserved LV function, LV function was significantly depressed in the reduced LV group, LVEF 55.00% [53.75, 60.0] vs. 35.00% [30.00, 39.25] (p < 0.001). Septum thickness, aortic valve area (AVA), and mean gradient across the aortic valve were significantly higher in the preserved LV function group, 14.00 mm [13.00, 15.0] vs. 12.50 mm [12.00, 14.0] (p < 0.001), 0.70 cm * [0.60, 0.80] vs. 0.65 cm * [0.50, 0.70] (p < 0.03) and 43.5 mmHg [32.25, 50.75] vs. 32.00 mmHg [27.00, 37.00] (p > 0.003). The main statistically significant difference between the two groups was a higher NLR at baseline in the reduced LV function group compared with the preserved LV function group, 2.85 [2.07, 4.78] vs. 3.90 [2.67, 5.26] (p < 0.04).

3.2. Procedural Data

Inflammatory marker dynamics: baseline total WBC, absolute cell counts of neutrophils, lymphocytes, and NLR and their dynamic changes after TAVI for the total study population are summarized in Table 2. In the entire 1:2 matched population, we noticed that there were significant kinetic changes in the WBC response (p value < 0.0001) from admission, to 24 h post procedure, to 72 h post procedure, with significant increases in WBC, neutrophils, and NLR, and significant decreases in absolute lymphocyte counts. Figure 2 shows the dynamic changes of white blood cells and the differential subsets in the entire study population at different times during the first 24 h and 72 h, and at six months post procedure. There was a significant increase in inflammatory markers, including total WBC, neutrophils, and NLR in all the study 1:2 matched population. When comparing the two subgroups as shown in Table 3 and Figure 3, patients with reduced left ventricular function had a significantly more pronounced inflammatory response at baseline. However, six months after TAVI, there was a similar extent of reduction in the inflammatory markers to about the same level of the in-patients, with reduced NLR compared to preserved ejection fraction, NLR 2.94 [2.01, 388] vs. 3.30 [2.06, 5.35], p = 0.288. NLR reduction in the low EF group from baseline to 6 months post procedure did not achieve statistical significance, 3.90 [2.67, 5.26] vs. 3.30 [2.06, 5.35] (p = 0.32).
Table 2. Dynamic changes of WBC and their subpopulations after TAVI of 1:2 matched population.
Table 2. Dynamic changes of WBC and their subpopulations after TAVI of 1:2 matched population.
Admission24 h72 h6 Monthsp1p2p3
WBC (K/μL)7.46 ± 2.2610.08 ± 3.559 ± 2.917.47 ± 2.38<0.0001<0.00011
Absolute neutrophils (K/μL)4.97 ± 2.068.19 ± 3.436.73 ± 2.794.89 ± 2.04<0.0001<0.00011
Absolute lymphocytes (K/μL)1.67 ± 1.11.1 ± 0.761.29 ± 0.591.76 ± 1.09<0.0001<0.00011
NLR 3.72 ± 2.89.76 ± 7.296.52 ± 4.663.36 ± 2.23<0.0001<0.00011
Values are mean ± SD; p1 = Comparison of pre-procedural values with those at 24 h; p2 = Comparison of pre-procedural values with those at 72 h; p3 = Comparison of pre-procedural values with those at 6 months.
Table 3. Timeline of dynamic changes of WBCs and its components of 1:2 matched population.
Table 3. Timeline of dynamic changes of WBCs and its components of 1:2 matched population.
OverallEF ≥ 50EF ≤ 40p Value
N = 156N = 104N = 52
Admission
WBC (K/μL) 7.10 [5.89, 8.43]7.06 [5.90, 8.14]7.25 [5.68, 8.90]0.565
Absolute neutrophils (K/μL)4.70 [3.51, 6.11]4.59 [3.52, 5.93]4.88 [3.48, 6.60]0.316
Absolute lymphocytes (K/μL)1.31 [1.10, 1.90]1.40 [1.14, 1.90]1.28 [1.00, 1.78]0.215
NLR 3.21 [2.22, 5.00]2.85 [2.07, 4.78]3.90 [2.67, 5.26]0.04
24 h post-procedure
WBC (K/μL) 9.53 [7.40, 11.80]9.65 [7.57, 11.88]9.10 [6.88, 11.10]0.371
Absolute neutrophils (K/μL) 7.65 [5.50, 9.46]7.70 [5.50, 9.58]7.60 [4.97, 9.40]0.434
Absolute lymphocytes (K/μL) 0.95 [0.66, 1.30]1.00 [0.70, 1.30]0.90 [0.60, 1.20]0.421
NLR7.83 [5.48, 12.07]7.40 [5.48, 12.18]8.35 [5.48, 10.47]0.678
72 h post-procedure
WBC (K/μL)8.49 [7.10, 10.38]8.55 [7.10, 10.40]8.30 [7.10, 9.70]0.497
Absolute neutrophils (K/μL)6.40 [4.83, 8.16]6.52 [4.88, 8.22]6.00 [4.83, 7.65]0.436
Absolute lymphocytes (K/μL)1.10 [0.80, 1.49]1.10 [0.88, 1.49]1.00 [0.80, 1.48]0.468
NLR5.71 [3.77, 8.09]5.65 [3.82, 8.15]6.16 [3.48, 8.01]0.863
6 months post-procedure
WBC (K/μL)7.10 [5.84, 8.65]7.05 [5.72, 8.24]7.20 [6.00, 9.30]0.38
Absolute neutrophils (K/μL)4.60 [3.40, 5.95]4.50 [3.40, 5.40]4.60 [3.55, 6.55]0.288
Absolute lymphocytes (K/μL) 1.60 [1.15, 1.92]1.60 [1.17, 2.08]1.60 [1.20, 1.86]0.844
NLR3.00 [2.02, 4.35]2.94 [2.01, 3.88]3.30 [2.06, 5.35]0.288
Values are presented as: median (ranges). Abbreviations: WBC = white blood cells; NLR = neutrophils to lymphocytes ratio. In a variable analysis for factors that may influence inflammatory markers post-TAVI for the whole study population, we found significant differences only in the procedure duration and contrast volume injected (Table 4).
Table 4. Variable analysis of factors that may influence inflammatory markers post-TAVI for the total population.
Table 4. Variable analysis of factors that may influence inflammatory markers post-TAVI for the total population.
Post-Procedure 24 h—WBCPost-Procedure 24 h—NEUT AbsPost-Procedure 24 h—LYMP AbsNLR 24 h
Median (IQR)p-ValueMedian (IQR)p-ValueMedian (IQR)p-ValueMedian (IQR)p-Value
Total9.53 (7.4–11.8) 7.65 (5.5–9.51) 0.95 (0.62–1.3) 7.83 (5.46–12.14)
Genderfemales9.6 (7.15–11.41)0.6347.77 (5.5–9.4)0.9520.9 (0.6–1.3)0.7368.02 (5.46–11.5)0.696
males9.4 (7.4–12.4)7.6 (5.5–10.1)1 (0.7–1.3)7.54 (5.5–12.36)
Age *, years 0.0530.511−0.0010.993−0.0420.6040.0170.829
BMI *, kg/m2 0.1120.1650.0970.2290.161 0.046−0.0390.627
HTNno8.7 (7.53–13.4)0.8606.9 (5.5–12)0.9750.8 (0.6–1.2)0.52110.46 (5.28–17.25)0.411
yes9.63 (7.15–11.8)7.685 (5.5–9.455)0.975 (0.66–1.3)7.63 (5.48–11.41)
Diabetesno9.67 (7.15–11.87)0.9567.67 (5.4–9.67)0.9440.94 (0.7–1.3)0.8897.66 (5.18–12.14)0.446
yes9.53 (7.6–11.67)7.625 (6.17–9.2)0.975 (0.6–1.3)7.87 (5.87–11.33)
AFno9.4 (7.53–11.87)0.6137.7 (5.7–9.6)0.2510.9 (0.7–1.3)0.2728.13 (5.62–12.77)0.075
yes9.685 (7.03–11)7 (5.1–9.2)1.035 (0.7–1.3)6.91 (4.78–10)
CADno9.45 (7.515–11.555)0.9947 (5.5–9.4)0.7690.94 (0.7–1.3)0.8537.54 (5.18–12.2)0.856
yes9.63 (7–11.87)7.7 (5.3–9.51)1 (0.6–1.3)7.83 (5.78–10.85)
PADno9.565 (7.45–12.085)0.4527.67 (5.5–9.79)0.3660.95 (0.6–1.3)0.8657.9 (5.26–12.77)0.486
yes9.415 (6.85–11)7.425 (5.05–9)0.935 (0.7–1.25)7.2 (5.73–9.64)
Baseline echo
LVEF (%) *0.1460.0700.1480.0660.0360.6560.0510.532
Septum thickness (mm) *0.1320.1020.1420.079−0.1370.0910.200 0.013
Procedure related
TAVI typesSEV9.8 (7.7–12.7)0.0767.7 (5.9–10.3)0.0561 (0.7–1.3)0.8487.85 (5.55–12.16)0.389
BEV9.25 (7.015–10.9)7.4 (5.11–8.95)0.975 (0.6–1.3)7.29 (5.27–10.47)
Contrast volume (mL) *0.1500.0620.162 0.044−0.0310.7050.188 0.019
Time (minutes) *0.1130.1650.166 0.041−0.191 0.0180.275 0.001
Post-dilatationno9.615 (7.53–11.8)0.5857.7 (5.5–9.4)0.7021 (0.7–1.3)0.4507.6 (5.46–12)0.749
yes9.15 (7–11.87)6.92 (5.5–9.6)0.855 (0.6–1.2)8.69 (5.5–12.14)
Data are presented as: median (ranges). Continuous variables between the various study groups were tested for normality by a Shapiro–Wilk test and when abnormal distribution was found, a Mann–Whitney test was performed. When the distribution was normal, a t-test was used. * Correlation between two continuous variables were tested by a Pearson’s correlation. Abbreviations: TAVI = transcatheter valve implantation; BMI = Body mass index; AF = atrial fibrillation; CAD = coronary artery disease; PAD = peripheral arterial disease; LVEF = left ventricle ejection fraction; SEV = self-expandable valve; BEV = balloon-expandable valve; WBC = white blood cells; Abs = absolute; NLR = neutrophils to lymphocytes ratio.

3.3. Clinical Outcomes

The thirty-day clinical outcomes according to the VARC-2 criteria are summarized in Table 5. We found no differences between the two subgroups according to MACE and mortality. The Kaplan–Meier survival curve at the four-year follow-up is shown in Figure 4. During the ten-year study period, we found no significant differences between the two groups according to the ejection fraction (p = 0.35).

4. Discussion

We have presented a matched-pair analysis comparing inflammatory biomarker behavior in patients who underwent TAVI for symptomatic severe aortic stenosis in two different cohorts: patients with reduced LV function versus patients with preserved LV function. The major findings of our study were: (i) the enhanced inflammatory state in patients with reduced LV function compared with preserved LV function is reflected by a higher baseline NLR; (ii) the effect of the TAVI on the reduction of the NLR inflammatory marker in the reduced LV group caught up with the NLR in the preserved LV function group after six months of follow-up; (iii) there are similarly favorable clinical outcomes, independent of the baseline LV function.
Inflammation contributes to the pathogenesis and progression of heart failure (HF) [12] as well as degenerative aortic stenosis [20,21]. Elevated inflammatory biomarkers, including NLR, were reported in the TIME-CHF study [9] that enrolled elderly patients with HF and severe aortic stenosis who were candidates for surgical replacement [22,23,24]. Our findings of elevated NLR in patients with severe aortic stenosis and reduced LV function are concordant with the publication of Avci et al. [22], who found higher NLR in patients presenting with these two entities. In our study, we added another parameter when analyzing the effect of TAVI on the inflammatory profile six months after the procedure, which mirrors complete resolution from inflammation-related features associated with the procedure.
Our findings are in agreement with the recent publication of Baratchi et al. [25], who found an anti-inflammatory effect of TAVI reflected by a significant reduction in the NLR. One possible pathophysiologic explanation is that the reduction of NLR after TAVI in patients with reduced LV function may be related to the effect of TAVI in decreasing hemodynamic resistance, which lowers the high shear stress that activates and perpetuates the inflammatory state, as experimentally demonstrated in the Baratchi publication [25].
Other significant baseline characteristic differences found were septum thickness, aortic valve area, and mean aortic valve gradient. These issues reflect the underling pathophysiology of patients with reduced LV function, which are superimposed with significant aortic stenosis activated compensatory pathways (i.e., chamber remodeling). These parameters are not statistically significant in the variable analysis (Table 4).
Finally, as described in the pivotal PARTNER study [5] and other studies [5,6,26], we show that there are no differences in the long-term mortality rates between patients with decreased or preserved LV function after TAVI. The apparent normalization of the inflammatory markers by TAVI, which was more pronounced in the patients with baseline reduced EF, may be associated with the observation that despite the presence of preexisting LV dysfunction, these patients do not have a poorer short-term outcome. However, this should be tested in larger cohorts and for extended periods of follow-up.
To the best of our knowledge, this is the first report that compares the inflammatory state behavior and kinetics in patients undergoing TAVI with low ejection fraction versus those with preserved LV function. In conclusion, we found that TAVI has a more pronounced anti-inflammatory effect in patients with a low baseline ejection fraction, and this effect may partly account for the beneficial clinical response, which is translated to similar outcomes when compared to patients with preserved LV function.

Author Contributions

Study concept and design: H.A.K., J.G. and A.B.; acquisition of data: H.A.K., G.G. and O.A.; analysis and interpretation of data: H.A.K., O.A. and A.B.; drafting of the manuscript: H.A.K., J.G. and A.B.; critical revision of the manuscript: G.G., L.P., M.J., O.P., S.S., S.G. and V.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Ethics Committee, Kaplan Medical Center (confirmation 0091-20-KMC).

Informed Consent Statement

Individual patient consent was waived because of the study’s retrospective design and the data collection from routine care.

Data Availability Statement

All relevant data are included within the manuscript. All other data presented in this study are available upon request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Halkos, M.E.; Chen, E.P.; Sarin, E.L.; Kilgo, P.; Thourani, V.H.; Lattouf, O.M.; Vega, J.D.; Morris, C.D.; Vassiliades, T.; Cooper, W.A. Aortic valve replacement for aortic stenosis in patients with left ventricular dysfunction. Ann. Thorac. Surg. 2009, 88, 746–751. [Google Scholar] [CrossRef]
  2. Pibarot, P.; Dumesnil, J.G. Low-flow, low-gradient aortic stenosis with normal and depressed left ventricular ejection fraction. J. Am. Coll. Cardiol. 2012, 60, 1845–1853. [Google Scholar] [CrossRef] [Green Version]
  3. Baron, S.J.; Arnold, S.V.; Herrmann, H.C.; Holmes, D.R.; Szeto, W.Y.; Allen, K.B.; Chhatriwalla, A.K.; Vemulapali, S.; O’Brien, S.; Dai, D. Impact of ejection fraction and aortic valve gradient on outcomes of transcatheter aortic valve replacement. J. Am. Coll. Cardiol. 2016, 67, 2349–2358. [Google Scholar] [CrossRef] [PubMed]
  4. Herrmann, H.C.; Pibarot, P.; Hueter, I.; Gertz, Z.M.; Stewart, W.J.; Kapadia, S.; Tuzcu, E.M.; Babaliaros, V.; Thourani, V.; Szeto, W.Y. Predictors of mortality and outcomes of therapy in low-flow severe aortic stenosis: A Placement of Aortic Transcatheter Valves (PARTNER) trial analysis. Circulation 2013, 127, 2316–2326. [Google Scholar] [CrossRef] [Green Version]
  5. Elmariah, S.; Palacios, I.F.; McAndrew, T.; Hueter, I.; Inglessis, I.; Baker, J.N.; Kodali, S.; Leon, M.B.; Svensson, L.; Pibarot, P. Outcomes of transcatheter and surgical aortic valve replacement in high-risk patients with aortic stenosis and left ventricular dysfunction: Results from the Placement of Aortic Transcatheter Valves (PARTNER) trial (cohort A). Circ. Cardiovasc. Interv. 2013, 6, 604–614. [Google Scholar] [CrossRef] [Green Version]
  6. Carreras, E.T.; Kaneko, T.; Ramirez-Del Val, F.; Pelletier, M.P.; Sobieszczyk, P.S.; Bhatt, D.L.; Shah, P.B. Impact of flow, gradient, and left ventricular function on outcomes after transcatheter aortic valve replacement. Catheter. Cardiovasc. Interv. 2018, 91, 798–805. [Google Scholar] [CrossRef]
  7. Passeri, J.J.; Elmariah, S.; Xu, K.; Inglessis, I.; Baker, J.N.; Alu, M.; Kodali, S.; Leon, M.B.; Svensson, L.G.; Pibarot, P. Transcatheter aortic valve replacement and standard therapy in inoperable patients with aortic stenosis and low EF. Heart 2015, 101, 463–471. [Google Scholar] [CrossRef]
  8. van der Boon, R.M.; Nuis, R.J.; Van Mieghem, N.M.; Benitez, L.M.; van Geuns, R.J.; Galema, T.W.; van Domburg, R.T.; Geleijnse, M.L.; Dager, A.; de Jaegere, P.P. Clinical outcome following transcatheter aortic valve implantation in patients with impaired left ventricular systolic function. Catheter. Cardiovasc. Interv. 2012, 79, 702–710. [Google Scholar] [CrossRef]
  9. Sanders-van Wijk, S.; Maeder, M.T.; Nietlispach, F.; Rickli, H.; Estlinbaum, W.; Erne, P.; Rickenbacher, P.; Peter, M.; Pfisterer, M.P.; Brunner-La Rocca, H.P. Long-Term Results of Intensified, N-Terminal-Pro-B-Type Natriuretic Peptide–Guided Versus Symptom-Guided Treatment in Elderly Patients With Heart Failure: Five-Year Follow-Up From TIME-CHF. Circ. Heart Fail. 2014, 7, 131–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. O’Connor, C.M.; Starling, R.C.; Hernandez, A.F.; Armstrong, P.W.; Dickstein, K.; Hasselblad, V.; Heizer, G.M.; Komajda, M.; Massie, B.M.; McMurray, J.J. Effect of nesiritide in patients with acute decompensated heart failure. N. Engl. J. Med. 2011, 365, 32–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Cho, J.H.; Cho, H.J.; Lee, H.Y.; Ki, Y.J.; Jeon, E.S.; Hwang, K.K.; Chae, S.C.; Baek, S.H.; Kang, S.M.; Choi, D.J. Neutrophil-lymphocyte ratio in patients with acute heart failure predicts in-hospital and long-term mortality. J. Clin. Med. 2020, 9, 557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Murphy, S.P.; Kakkar, R.; McCarthy, C.P.; Januzzi Jr, J.L. Inflammation in heart failure: JACC state-of-the-art review. J. Am. Coll. Cardiol. 2020, 75, 1324–1340. [Google Scholar] [CrossRef] [PubMed]
  13. Dick, S.A.; Epelman, S. Chronic heart failure and inflammation: What do we really know? Circ. Res. 2016, 119, 159–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sorysz, D.; Januszek, R.; Sowa-Staszczak, A.; Grochowska, A.; Opalińska, M.; Bagieński, M.; Zawiślak, B.; Dziewierz, A.; Tokarek, T.; Krawczyk-Ożóg, A. The Usefulness of [18F] F-Fluorodeoxyglucose and [18F] F-Sodium Fluoride Positron Emission Tomography Imaging in the Assessment of Early-Stage Aortic Valve Degeneration after Transcatheter Aortic Valve Implantation (TAVI)—Protocol Description and Preliminary Results. J. Clin. Med. 2021, 10, 431. [Google Scholar]
  15. Friedman, G.D.; Klatsky, A.L.; Siegelaub, A. The leukocyte count as a predictor of myocardial infarction. N. Engl. J. Med. 1974, 290, 1275–1278. [Google Scholar] [CrossRef]
  16. Horne, B.D.; Anderson, J.L.; John, J.M.; Weaver, A.; Bair, T.L.; Jensen, K.R.; Renlund, D.G.; Muhlestein, J.B.; Group IHCS. Which white blood cell subtypes predict increased cardiovascular risk? J. Am. Coll. Cardiol. 2005, 45, 1638–1643. [Google Scholar] [CrossRef] [Green Version]
  17. Duffy, B.K.; Gurm, H.S.; Rajagopal, V.; Gupta, R.; Ellis, S.G.; Bhatt, D.L. Usefulness of an elevated neutrophil to lymphocyte ratio in predicting long-term mortality after percutaneous coronary intervention. Am. J. Cardiol. 2006, 97, 993–996. [Google Scholar] [CrossRef]
  18. Azab, B.; Jaglall, N.; Atallah, J.P.; Lamet, A.; Raja-Surya, V.; Farah, B.; Lesser, M.; Widmann, W.D. Neutrophil-lymphocyte ratio as a predictor of adverse outcomes of acute pancreatitis. Pancreatology 2011, 11, 445–452. [Google Scholar] [CrossRef]
  19. Uthamalingam, S.; Patvardhan, E.A.; Subramanian, S.; Ahmed, W.; Martin, W.; Daley, M.; Capodilupo, R. Utility of the neutrophil to lymphocyte ratio in predicting long-term outcomes in acute decompensated heart failure. Am. J. Cardiol. 2011, 107, 433–438. [Google Scholar] [CrossRef]
  20. Im Cho, K.; Sakuma, I.; Sohn, I.S.; Jo, S.H.; Koh, K.K. Inflammatory and metabolic mechanisms underlying the calcific aortic valve disease. Atherosclerosis 2018, 277, 60–65. [Google Scholar] [CrossRef] [PubMed]
  21. Dweck, M.R.; Khaw, H.; Sng, G.K.; Luo, E.; Baird, A.; Williams, M.C.; Makiello, P.; Mirsadraee, S.; Joshi, N.; van Beek, E. Aortic stenosis, atherosclerosis, and skeletal bone: Is there a common link with calcification and inflammation? Eur. Heart J. 2013, 34, 1567–1574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ari, M.E.; Ekici, F.; Çetin, İİ.; Tavil, E.B.; Yaralı, N.; Işık, P.; Hazırolan, T.; Tunç, B. Assessment of left ventricular functions and myocardial iron load with tissue Doppler and speckle tracking echocardiography and T2* MRI in patients with β-thalassemia major. Echocardiography 2017, 34, 383–389. [Google Scholar] [CrossRef]
  23. Cho, K.I.; Cho, S.H.; Her, A.Y.; Singh, G.B.; Shin, E.S. Prognostic utility of neutrophil-to-lymphocyte ratio on adverse clinical outcomes in patients with severe calcific aortic stenosis. PloS ONE 2016, 11, e0161530. [Google Scholar] [CrossRef] [PubMed]
  24. Song, J.; Zheng, Q.; Ma, X.; Zhang, Q.; Xu, Z.; Zou, C.; Wang, Z. Predictive roles of neutrophil-to-lymphocyte ratio and C-reactive protein in patients with calcific aortic valve disease. Int. Heart J. 2019, 60, 345–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Baratchi, S.; Zaldivia, M.T.; Wallert, M.; Loseff-Silver, J.; Al-Aryahi, S.; Zamani, J.; Thurgood, P.; Salim, A.; Htun, N.M.; Stub, D. Transcatheter Aortic Valve Implantation Represents an Anti-Inflammatory Therapy Via Reduction of Shear Stress–Induced, Piezo-1–Mediated Monocyte Activation. Circulation 2020, 142, 1092–1105. [Google Scholar] [CrossRef]
  26. Chieffo, A.; Buchanan, G.L.; Van Mieghem, N.M.; Tchetche, D.; Dumonteil, N.; Latib, A.; Van Der Boon, R.M.; Vahdat, O.; Marcheix, B.; Farah, B. Transcatheter aortic valve implantation with the Edwards SAPIEN versus the Medtronic CoreValve Revalving system devices: A multicenter collaborative study: The PRAGMATIC Plus Initiative (Pooled-RotterdAm-Milano-Toulouse In Collaboration). J. Am. Coll. Cardiol. 2013, 61, 830–836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Study flowchart of patients from 2010 to 2019. A total of 370 patients were treated with TAVI. After excluding 56 patients and doing propensity score matching, a total of 156 patients were finally included in the analysis.
Figure 1. Study flowchart of patients from 2010 to 2019. A total of 370 patients were treated with TAVI. After excluding 56 patients and doing propensity score matching, a total of 156 patients were finally included in the analysis.
Jcm 10 04148 g001
Figure 2. Dynamic changes of leukocytes and its components after TAVI for the matched population, (A) dynamic changes in total WBC with time, (B) dynamic changes in Neutrophils with time, (C) dynamic changes of lymphocytes with time, (D) dynamic changes of NL ratio with time. LYMP. abs = Absolute Lymphocytes (K/μL); NEUT. abs = Absolute Neutrophils (K/μL); WBC = White Blood Cells (K/μL); NLR = Neutrophils to Lymphocytes Ratio; ns = not significant; **** = significant (p value < 0.01), Dots are the values beyond the extreme line that shows potential outliers.
Figure 2. Dynamic changes of leukocytes and its components after TAVI for the matched population, (A) dynamic changes in total WBC with time, (B) dynamic changes in Neutrophils with time, (C) dynamic changes of lymphocytes with time, (D) dynamic changes of NL ratio with time. LYMP. abs = Absolute Lymphocytes (K/μL); NEUT. abs = Absolute Neutrophils (K/μL); WBC = White Blood Cells (K/μL); NLR = Neutrophils to Lymphocytes Ratio; ns = not significant; **** = significant (p value < 0.01), Dots are the values beyond the extreme line that shows potential outliers.
Jcm 10 04148 g002
Figure 3. Difference in two matched subgroups (EF ≥ 50 vs. EF ≤ 40) for leukocytes and its components after TAVI, (A) difference in total WBC with time, (B) difference in Neutrophils with time, (C) difference in lymphocytes with time, (D) difference in NL ratio with time. LYMP. abs = Absolute Lymphocytes (K/μL); NEUT. abs = Absolute Neutrophils (K/μL); WBC = White Blood Cells (K/μL); NLR = Neutrophils to Lymphocytes Ratio, Dots are the values beyond the extreme line that shows potential outliers.
Figure 3. Difference in two matched subgroups (EF ≥ 50 vs. EF ≤ 40) for leukocytes and its components after TAVI, (A) difference in total WBC with time, (B) difference in Neutrophils with time, (C) difference in lymphocytes with time, (D) difference in NL ratio with time. LYMP. abs = Absolute Lymphocytes (K/μL); NEUT. abs = Absolute Neutrophils (K/μL); WBC = White Blood Cells (K/μL); NLR = Neutrophils to Lymphocytes Ratio, Dots are the values beyond the extreme line that shows potential outliers.
Jcm 10 04148 g003
Figure 4. Kaplan Meir curve based on all cause mortality for patients underwent TAVI, showing no difference in survival between the two TAVI matched subgroups according to ejection fraction, LVEF = Left Ventricular Ejection Fraction.
Figure 4. Kaplan Meir curve based on all cause mortality for patients underwent TAVI, showing no difference in survival between the two TAVI matched subgroups according to ejection fraction, LVEF = Left Ventricular Ejection Fraction.
Jcm 10 04148 g004
Table 1. Baseline characteristics of the 1:2 matched population.
Table 1. Baseline characteristics of the 1:2 matched population.
OverallEF ≥ 50EF ≤ 40p Value
Clinical characteristicN = 156N = 104N = 52
Age, years81.00 [76.00, 85.00]81.50 [74.75, 86.25]81.00 [77.00, 85.00]0.737
Male (%)89 (57%)60 (57.7%)29 (55.8%)0.954
Body mass index, kg/m 26.89 [23.44, 29.33]26.33 [22.72, 30.55]27.05 [24.08, 29.11]0.506
Hypertension (%)145 (93%)97 (93.3%)48 (92.3%)1
Diabetes (%)67 (43%)44 (42.3%)23 (44.2%)0.954
Dyslipidemia (%)118 (76%)75 (72.1%)43 (82.7%)0.21
Smoker (%)21 (13%)14 (13.5%)7 (13.5%)1
Atrial fibrillation (%)50 (32%)35 (34.0%)15 (30.0%)0.758
Coronary artery disease (%)71 (46%)42 (40.8%)29 (55.8%)0.11
Peripheral artery disease (%)32 (21%)16 (15.4%)16 (30.8%)0.042
Previous myocardial infarction (%)21 (13%)10 (9.6%)11 (21.6%)0.073
Previous stroke (%)20 (13%)14 (13.5%)6 (11.5%)0.933
Previous pacemaker (%)22 (14%)14 (13.5%)8 (15.4%)0.935
CABG (%)12 (0.7%)5 (7.9%)7 (17.1%)0.266
STS Score8.01 [5.1, 10.3]8.04 [4.8, 10.2]8.3 [5.2, 10.6]0.617
LVEF (%)52.50 [39.75, 60.00]55.00 [53.75, 60.0]35.00 [30.00, 39.25]<0.001
Septum thickness (mm)13.00 [12.00, 15.00]14.00 [13.00, 15.0]12.50 [12.00, 14.0]0.001
Aortic valve area (cm)0.70 [0.60, 0.80]0.70 [0.60, 0.80]0.65 [0.50, 0.70]0.035
Aortic valve gradient- mean (mm Hg)37.00 [29.50, 47.00]43.50 [32.25, 50.75]32.00 [27.00, 37.00]0.003
Contrast volume (mL)120.00 [90.00, 157.0]136.44 (63.55)122.04 (51.51)0.158
Time (minutes)84.00 [68.00, 105.75]93.44 (33.77)84.23 (32.07)0.106
WBC (K/μL)7.10 [5.89, 8.43]7.06 [5.90, 8.14]7.25 [5.68, 8.90]0.565
Absolute Neutrophils (K/μL)4.70 [3.51, 6.11]4.59 [3.52, 5.93]4.88 [3.48, 6.60]0.316
Absolute Lymphocytes (K/μL)1.31 [1.10, 1.90]1.28 [1.00, 1.78]1.40 [1.14, 1.90]0.215
NLR3.21 [2.22, 5.00]2.85 [2.07, 4.78]3.90 [2.67, 5.26]0.04
Values are presented as: median (ranges) or n (%). Abbreviations: CABG = coronary artery bypass; STS = Society of Thoracic Surgeons; LVEF = left ventricle ejection fraction; WBC = white blood cells; NLR = neutrophils to lymphocytes ratio.
Table 5. Thirty-day outcomes of patients after TAVI of 1:2 matched population .
Table 5. Thirty-day outcomes of patients after TAVI of 1:2 matched population .
VariableTotal EF ≥ 50 EF ≤ 40p Value
Mortality8 (5.1%)5 (4.8%)3 (5.7%)0.271
Bleeding8 (0.5%)6 (5.8%)2 (3.8%)0.898
Major vascular complication13 (0.8%)9 (8.7%)4 (7.7%)1
Stroke3 (0.1%)1 (1.0%)2 (3.8%)0.536
Myocardial infarction3 (0.1%)2 (1.9%)1 (1.9%)1
Acute kidney injury7 (0.5%)4 (3.8%)3 (5.8%)0.891
Arrhythmia42 (27%)29 (27.9%)13 (25.0%)0.848
† Categorical and nominal variables were reported by prevalence and percentages and were analyzed by Pearson’s chi-square (χ²) test and Fisher’s exact test.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abu Khadija, H.; Gandelman, G.; Ayyad, O.; Poles, L.; Jonas, M.; Paz, O.; Goland, S.; Shimoni, S.; Meledin, V.; George, J.; et al. Comparative Analysis of the Kinetic Behavior of Systemic Inflammatory Markers in Patients with Depressed versus Preserved Left Ventricular Function Undergoing Transcatheter Aortic Valve Implantation. J. Clin. Med. 2021, 10, 4148. https://doi.org/10.3390/jcm10184148

AMA Style

Abu Khadija H, Gandelman G, Ayyad O, Poles L, Jonas M, Paz O, Goland S, Shimoni S, Meledin V, George J, et al. Comparative Analysis of the Kinetic Behavior of Systemic Inflammatory Markers in Patients with Depressed versus Preserved Left Ventricular Function Undergoing Transcatheter Aortic Valve Implantation. Journal of Clinical Medicine. 2021; 10(18):4148. https://doi.org/10.3390/jcm10184148

Chicago/Turabian Style

Abu Khadija, Haitham, Gera Gandelman, Omar Ayyad, Lion Poles, Michael Jonas, Offir Paz, Sorel Goland, Sara Shimoni, Valery Meledin, Jacob George, and et al. 2021. "Comparative Analysis of the Kinetic Behavior of Systemic Inflammatory Markers in Patients with Depressed versus Preserved Left Ventricular Function Undergoing Transcatheter Aortic Valve Implantation" Journal of Clinical Medicine 10, no. 18: 4148. https://doi.org/10.3390/jcm10184148

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop