Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Clin Auton Res ; 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39312106

ABSTRACT

PURPOSE: Autonomic dysregulation is observed in heart failure (HF) with reduced ejection fraction (HFrEF). Abnormal heart rate variability (HRV), a measure of such dysregulation, is associated with poor prognosis in HFrEF. It is unknown if novel HRV metrics normalize in the patients with recovered ejection fraction (HFrecEF) compared to persistent HFrEF. The aim of this study was to investigate novel HRV indexes in persistent HFrEF in comparison to HFrecEF METHODS: A standard 10-min electrocardiography measurement was performed in patients categorized in four groups: persistent HFrEF (n = 40), HFrecEF (n = 41), stage A HF (n = 73) and healthy controls (n = 40). RESULTS: All HRV indexes were significantly different between the four groups. Specifically, novel metrics, such as higher parasympathetic nervous system (PNS) index and lower sympathetic nervous system (SNS) index, were observed in the HFrecEF group compared to the persistent HFrEF group. In multiple logistic regression analysis, higher PNS index (odds ratio [OR] 2.02, 95% confidence interval [CI] 1.17-3.49; p = 0.01) and lower SNS index (OR 0.68, 95% CI 0.52-0.87; p = 0.002) were associated with HFrecEF. Receiver operating characteristic analysis showed that the SNS index had the highest area under the curve (AUC), followed by the PNS index and mean heart rate for the HF phenotype regarding EF recovery (AUC = 0.71, 0.69 and 0.69, respectively). CONCLUSION: Myocardial functional recovery in HFrEF is associated with improved parasympathetic activity and reduced sympathetic activity, as reflected in the PNS and SNS indexes. These novel metrics can be potentially used to aid in identifying recovered versus non-recovered phenotypes in patients with HFrEF.

2.
Hypertens Res ; 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261699

ABSTRACT

Metabolic syndrome (MetS) induces a systemic inflammatory state which can lead to cardiomyopathy, manifesting clinically as heart failure (HF) with preserved ejection fraction (HFpEF). MetS components are intricately linked to the pathophysiologic processes of myocardial remodeling. Increased sympathetic nervous system activity, which is noted as an upstream factor of MetS, has been linked to adverse myocardial structural changes. Since renal denervation and vagus nerve stimulation have a sympathoinhibitory effect, attention has been paid to the cardioprotective effects of autonomic neuromodulation. In this review, the pathophysiology underlying the relationship between MetS and HF is elucidated, and the evidence regarding autonomic neuromodulation in HFpEF is summarized.

3.
Respir Physiol Neurobiol ; 328: 104313, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39122159

ABSTRACT

INTRODUCTION: The interaction between the cardiovascular and respiratory systems in healthy subjects is determined by the autonomic nervous system and reflected in respiratory sinus arrhythmia. Recently, another pattern of cardio-respiratory coupling (CRC) has been proposed linking synchronization of heart and respiratory system. However, CRC has not been studied precisely in heart failure (HF) with reduced ejection fraction (EF) (HFrEF) according to the myocardial recovery. METHODS: 10-min resting electrocardiography measurements were performed in persistent HFrEF patients (n=40) who had a subsequent left ventricular EF (LVEF) of ≤ 40 %, HF with recovered EF patients (HFrecEF) (n=41) who had a subsequent LVEF of > 40 % and healthy controls (n=40). Respiratory frequency, respiratory rate, CRC index, time-domain, frequency-domain and nonlinear heart rate variability indices were obtained using standardized software-Kubios™. CRC index was defined as respiratory high-frequency peak minus heart rate variability high-frequency peak. RESULTS: Respiratory rate was positively correlated with high-frequency (HF) peak (Hz) in both persistent HFrEF group (p<0.001) and HFrecEF group (p<0.001), while respiratory rate was negatively correlated with HF power (ms2) in the healthy controls (p<0.05). CRC index was lowest in the persistent HFrEF group followed by HFrecEF and was high in healthy controls (0.008 vs 0.012 vs 0.056 Hz, p=0.03). CONCLUSION: CRC index was lowest in patients with impaired myocardial recovery, which indicates that cardio-respiratory synchrony is stronger in persistent HFrEF. This may represent a higher HF peak (Hz)/lower HF power (ms2) and abnormal sympathovagal balance in persistent HFrEF group compared to healthy controls. Further work is underway to tests this hypothesis and determine the utility of CRC index in HF phenotypes and its utility as a potential biomarker of response with neuromodulation.


Subject(s)
Electrocardiography , Heart Failure , Heart Rate , Stroke Volume , Humans , Heart Failure/physiopathology , Male , Female , Middle Aged , Stroke Volume/physiology , Aged , Heart Rate/physiology , Recovery of Function/physiology , Heart/physiopathology , Respiratory Rate/physiology
5.
Article in English | MEDLINE | ID: mdl-38969912

ABSTRACT

Higher blood pressure (BP) variability (BPV) was shown to be strong predictors of poor cardiovascular outcomes in heart failure (HF). It is currently unknown if low-level tragus stimulation (LLTS) would lead to improvement in BPV in acute HF (AHF). The 22 patients with AHF (median 80 yrs, males 60%) were randomly assigned to active or sham group using an ear clip attached to the tragus (active group) or the earlobe (sham group) for 1 h daily over 5 days. In the active group, standard deviation (SD), coefficient of variation (CV) and δ in SBP were significantly decreased after LLTS (all p < 0.05). All the changes in SD, CV and δ in SBP before and after stimulation were also significantly different between active and sham groups (all p < 0.05). This proof-of-concept study demonstrates the beneficial effects of LLTS on BPV in AHF.

6.
Card Electrophysiol Clin ; 16(3): 307-314, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084723

ABSTRACT

The autonomic nervous system plays a central role in the pathogenesis of arrhythmias. Preclinical and clinical studies have demonstrated the therapeutic effect of neuromodulation at multiple anatomic targets across the neurocardiac axis for the treatment of arrhythmias. In this review, we discuss the rationale and clinical application of noninvasive neuromodulation techniques in treating arrhythmias and explore associated barriers and future directions, including optimization of stimulation parameters and patient selection.


Subject(s)
Arrhythmias, Cardiac , Humans , Arrhythmias, Cardiac/therapy , Arrhythmias, Cardiac/physiopathology , Autonomic Nervous System/physiopathology , Electric Stimulation Therapy/methods
7.
Heart Rhythm ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848857

ABSTRACT

Diabetes mellitus (DM) confers an increased risk of sudden cardiac death (SCD) independent of its associated cardiovascular comorbidities. DM induces adverse structural, electrophysiologic, and autonomic cardiac remodeling that can increase one's risk of ventricular arrhythmias and SCD. Although glycemic control and prevention of microvascular and macrovascular complications are cornerstones in the management of DM, they are not adequate for the prevention of SCD. In this narrative review, we describe the contribution of DM to the pathophysiologic mechanism of SCD beyond its role in atherosclerotic cardiovascular disease and heart failure. On the basis of this pathophysiologic framework, we outline potential preventive and therapeutic strategies to mitigate the risk of SCD in this population of high-risk patients.

8.
Am J Clin Oncol ; 47(9): 425-430, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38800981

ABSTRACT

OBJECTIVES: Cardiotoxic chemotherapy is used to treat malignancies such as breast cancer and lymphoma. These treatments predispose patients to cardiotoxicity that can lead to cancer treatment-related cardiac dysfunction (CTRCD). The use of high doses of anthracyclines or in combination with human epidermal growth factor receptor 2 antagonists is associated with a progressively higher risk of CTRCD. CTRCD is preceded by increased activation of the sympathetic nervous system and abnormal left ventricular mechanical deformation as measured by abnormal global longitudinal strain (GLS). Low-level tragus stimulation (LLTS) is a new, safe, noninvasive technique that offers great potential to reduce increased sympathetic activation and improve GLS. Here, we describe a study method to examine the effects of LLTS on autonomic balance and cardiac function in breast cancer or lymphoma patients treated with anthracyclines. METHODS: A first-in-human pilot, randomized, double-blind feasibility study will evaluate 104 patients (age >50 y) with breast cancer or lymphoma who receive anthracyclines with one additional CTRCD risk factor. Patients undergo 2 weeks of LLTS daily (1 h/d). Autonomic balance will be measured using heart rate variability metrics. Strain imaging using GLS will be performed pre and post-LLTS. Endothelial inflammation and oxidative stress measures will be performed using in vitro assays at baseline and after 2 weeks. CONCLUSION: We hypothesize that LLTS stabilizes sympathovagal imbalance and improves cardiac performance in anthracycline-treated patients with breast cancer or lymphoma.


Subject(s)
Breast Neoplasms , Cardiotoxicity , Humans , Pilot Projects , Female , Cardiotoxicity/etiology , Breast Neoplasms/drug therapy , Double-Blind Method , Middle Aged , Lymphoma/drug therapy , Anthracyclines/adverse effects , Feasibility Studies , Male , Cardiovascular Diseases/chemically induced
10.
J Am Heart Assoc ; 13(3): e032100, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38258658

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) increases risk of embolic stroke, and in postoperative patients, increases cost of care. Consequently, ECG screening for AF in high-risk patients is important but labor-intensive. Artificial intelligence (AI) may reduce AF detection workload, but AI development presents challenges. METHODS AND RESULTS: We used a novel approach to AI development for AF detection using both surface ECG recordings and atrial epicardial electrograms obtained in postoperative cardiac patients. Atrial electrograms were used only to facilitate establishing true AF for AI development; this permitted the establishment of an AI-based tool for subsequent AF detection using ECG records alone. A total of 5 million 30-second epochs from 329 patients were annotated as AF or non-AF by expert ECG readers for AI training and validation, while 5 million 30-second epochs from 330 different patients were used for AI testing. AI performance was assessed at the epoch level as well as AF burden at the patient level. AI achieved an area under the receiver operating characteristic curve of 0.932 on validation and 0.953 on testing. At the epoch level, testing results showed means of AF detection sensitivity, specificity, negative predictive value, positive predictive value, and F1 (harmonic mean of positive predictive value and sensitivity) as 0.970, 0.814, 0.976, 0.776, and 0.862, respectively, while the intraclass correlation coefficient for AF burden detection was 0.952. At the patient level, AF burden sensitivity and positive predictivity were 96.2% and 94.5%, respectively. CONCLUSIONS: Use of both atrial electrograms and surface ECG permitted development of a robust AI-based approach to postoperative AF recognition and AF burden assessment. This novel tool may enhance detection and management of AF, particularly in patients following operative cardiac surgery.


Subject(s)
Atrial Fibrillation , Humans , Atrial Fibrillation/diagnosis , Artificial Intelligence , Electrophysiologic Techniques, Cardiac , Electrocardiography/methods , Hospitals
12.
Heart Rhythm ; 21(4): 464-470, 2024 04.
Article in English | MEDLINE | ID: mdl-38104955

ABSTRACT

Cardioneuroablation (CNA) is being increasingly used to treat patients with vasovagal syncope (VVS). Bradycardia, in the cardioinhibitory subtype of VVS, results from transient parasympathetic overactivity leading to sinus bradycardia and/or atrioventricular block. By mitigating parasympathetic overactivity, CNA has been shown to improve VVS symptoms in clinical studies with relatively small sample sizes and short follow-up periods (<5 years) at selected centers. However, CNA may potentially tip the autonomic balance to a state of sympathovagal imbalance with attenuation of cardiac parasympathetic activity. A higher heart rate is associated with adverse cardiovascular events and increased mortality in healthy populations without cardiovascular diseases. Chronic sympathovagal imbalance may also affect the pathophysiology of spectra of cardiovascular disorders including atrial and ventricular arrhythmias. This review addresses potential long-term pathophysiological consequences of CNA for VVS.


Subject(s)
Bradycardia , Syncope, Vasovagal , Humans , Syncope, Vasovagal/diagnosis , Syncope, Vasovagal/surgery , Arrhythmias, Cardiac , Heart Atria , Sick Sinus Syndrome
13.
J Innov Card Rhythm Manag ; 14(12): 5690-5696, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38155725

ABSTRACT

Pulmonary vein isolation via cryoballoon (CB) ablation is the cornerstone ablation strategy for the treatment of atrial fibrillation (AF). Acute intraprocedural hypotensive and/or bradycardic responses have been reported in patients undergoing CB ablation for AF. However, it remains unclear as to whether these are due to a true vagal response (VR), which can be used to predict long-term outcomes of CB ablation. We analyzed 139 freezes across 17 patients who received CB ablation for paroxysmal AF, measuring vital signs and freeze characteristics. Only one freeze was associated with both hypotension and bradycardia, constituting a true VR. Several freezes were associated with hypotension only that did not respond to atropine administration, suggesting that these responses are not associated with a VR. Hypotensive responses were significantly associated with ice bubble bursts during CB deflation. Unlike the true VR reported in patients undergoing conscious sedation, the presence of acute hypotension shortly after CB deflation cannot be used as a predictor for long-term ablation outcomes.

14.
Clin Auton Res ; 33(6): 767-775, 2023 12.
Article in English | MEDLINE | ID: mdl-37943335

ABSTRACT

PURPOSE: Acute decompensated heart failure (ADHF) is associated with inflammation, oxidative stress, and excess sympathetic drive. It is unknown whether neuromodulation would improve inflammation and oxidative stress in acute heart failure. We, therefore, performed this proof-of-concept study to evaluate the effects of neuromodulation using noninvasive low-level tragus stimulation on inflammation and oxidative stress in ADHF. METHODS: Nineteen patients with ejection fraction < 40% were randomized to neuromodulation 4 h twice daily (6-10 a.m. and 6-10 p.m.) (n = 8) or sham stimulation (n = 11) during hospital admission. All patients received standard-of-care treatment. Blood samples were collected at admission and discharge. Serum cytokines were assayed using standard immunosorbent techniques. Reactive oxygen species inducibility from cultured coronary endothelial cells exposed to patient sera was determined using a dihydrodichlorofluorescein probe test (expressed as fluorescein units). RESULTS: Compared to sham stimulation, neuromodulation was associated with a significant reduction of circulating serum interleukin-6 levels (-78% vs. -9%; p = 0.012). Similarly, neuromodulation led to a reduction of endothelial cell oxidative stress in the neuromodulation group (1363 units to 978 units, p = 0.003) compared to sham stimulation (1146 units to 1083 units, p = 0.094). No significant differences in heart rate, blood pressure, or renal function were noted between the two groups. CONCLUSION: In this proof-of-concept pilot study, in acute decompensated heart failure, neuromodulation was feasible and safe and was associated with a reduction in systemic inflammation and attenuation of coronary endothelial cellular oxidative stress. CLINICAL TRIAL REGISTRATION: NCT02898181.


Subject(s)
Endothelial Cells , Heart Failure , Humans , Pilot Projects , Heart Failure/therapy , Inflammation/therapy , Oxidative Stress
15.
Res Sq ; 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37790298

ABSTRACT

Purpose: Acute decompensated heart failure is associated with inflammation, oxidative stress, and excess sympathetic drive. It is unknown if neuromodulation would improve inflammation and oxidative stress in acute heart failure. We, therefore, performed this proof-of-concept study to evaluate the effects of neuromodulation using noninvasive low-level Tragus stimulation on inflammation and oxidative stress in ADHF. Methods: 19 patients with ejection fraction < 40% were randomized to neuromodulation- 4 hours twice daily (6 AM-10 AM and 6 PM-10 PM) (n = 8) or sham stimulation (n = 11) during hospital admission. All patients received standard-of-care treatment. Blood samples were collected at admission and discharge. Serum cytokines were assayed using standard immunosorbent techniques. Reactive oxygen species inducibility from cultured coronary endothelial cells exposed to patient sera was determined using dihydrodichlorofluorescein probe test (expressed as fluorescein units). Results: Compared to sham stimulation, neuromodulation was associated with a significant reduction of circulating serum Interleukin-6 levels (-78% vs -9%; p = 0.012). Similarly, neuromodulation led to reduction of endothelial cell oxidative stress, in the neuromodulation group (1363 units to 978 units, p = 0.003) compared to sham stimulation (1146 units to 1083 units, p = 0.094). No significant difference in heart rate, blood pressure or renal function were noted between the two groups. Conclusion: In this proof-of-concept pilot study, in acute systolic heart failure, neuromodulation was feasible and safe and was associated with a reduction in systemic inflammation and attenuation of cellular oxidative stress. Clinical trial: NCT02898181.

16.
Trials ; 24(1): 683, 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37872628

ABSTRACT

BACKGROUND: The autonomic nervous system can be responsible for the initiation and maintenance of arrhythmias. Low-level tragus stimulation (LLTS), a noninvasive form of autonomic neuromodulation, has been shown to be effective in treating atrial fibrillation. We intended to treat frequent premature ventricular complexes (PVCs) with LLTS. METHODS AND DESIGN: The present study will be a prospective multicenter, double-blind, randomized, controlled trial to assess the antiarrhythmic effects of LLTS on frequent PVCs in patients without structured heart disease (SHD). A total of 100 patients with PVC burden > 10% will be randomly assigned to the active or sham LLTS in 1:1 fashion and receive the proposed intervention for 6 months. The primary outcome is PVC burden at 6 months as assessed by 10 days of continuous ambulatory electrocardiographic monitoring. Secondary outcomes include heart rate variability (HRV), quality of life, skin sympathetic nerve activity, and inflammatory markers. Adverse events will also be recorded. DISCUSSION: The present trial will be the first to evaluate the effect of LLTS on frequent PVCs on patients without SHD. LLTS may serve as a low-cost, minimal-risk, and non-invasive alternative to conventional antiarrhythmic therapy. TRIAL REGISTRATION: ClinicalTrial.gov NCT04909528. Registered on 17 June 2021. World health organization trial registration data set was shown in Supplementary Table 1.


Subject(s)
Atrial Fibrillation , Vagus Nerve Stimulation , Ventricular Premature Complexes , Humans , Vagus Nerve Stimulation/adverse effects , Quality of Life , Prospective Studies , Ventricular Premature Complexes/diagnosis , Ventricular Premature Complexes/therapy , Atrial Fibrillation/diagnosis , Atrial Fibrillation/therapy , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
17.
Life Sci ; 331: 122069, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37666387

ABSTRACT

Mitigation of cardiac autonomic dysregulation by neuromodulation technologies is emerging as a new therapeutic modality of heart failure (HF). This recent progress has necessitated the identification of a biomarker for the quantification of sympathovagal balance, the potential target of 'neuromodulation' strategies. The currently available autonomic nervous system assessment parameters do not truly reflect the sympathovagal balance of the ventricle. Protein kinase A (PKA) is an intracellular enzyme that plays a major role in the pathophysiology of functional and structural ventricular remodeling in HF. Interestingly, sympathetic and parasympathetic activations exert reciprocal influence on the activity of PKA. The current review attempts to evaluate the potential concept and feasibility of using in vitro assessment of PKA activity as a marker of sympathovagal balance in HF.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Heart Failure , Humans , Heart , Autonomic Nervous System , Heart Ventricles
18.
Life Sci ; 333: 122122, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37774940

ABSTRACT

Abnormal cardiac metabolism or cardiac metabolic remodeling is reported before the onset of heart failure with reduced ejection fraction (HFrEF) and is known to trigger and maintain the mechanical dysfunction and electrical, and structural abnormalities of the ventricle. A dysregulated cardiac autonomic tone characterized by sympathetic overdrive with blunted parasympathetic activation is another pathophysiological hallmark of HF. Emerging evidence suggests a link between autonomic nervous system activity and cardiac metabolism. Chronic ß-adrenergic activation promotes maladaptive metabolic remodeling whereas cholinergic activation attenuates the metabolic aberrations through favorable modulation of key metabolic regulatory molecules. Restoration of sympathovagal balance by neuromodulation strategies is emerging as a novel nonpharmacological treatment strategy in HF. The current review attempts to evaluate the 'neuro-metabolic axis' in HFrEF and whether neuromodulation can mitigate the adverse metabolic remodeling in HFrEF.


Subject(s)
Heart Failure , Humans , Stroke Volume/physiology , Heart , Autonomic Nervous System , Cholinergic Agents
19.
JACC Clin Electrophysiol ; 9(7 Pt 2): 1196-1206, 2023 07.
Article in English | MEDLINE | ID: mdl-37086229

ABSTRACT

Simultaneous activation of the sympathetic and parasympathetic nervous systems is crucial for the initiation of paroxysmal atrial fibrillation (AF). However, unbalanced activation of the sympathetic system is characteristic of autonomic remodeling in long-standing persistent AF. Moreover, the adrenergic activation-induced metabolic derangements provide a milieu for acute AF and promote the transition from the paroxysmal to the persistent phase of AF. On the other hand, cholinergic activation ameliorates the maladaptive metabolic remodeling in the face of metabolic challenges. Selective inhibition of the sympathetic system and restoration of the balance of the cholinergic system by neuromodulation is emerging as a novel nonpharmacologic strategy for managing AF. This review explores the link between cardiac autonomic and metabolic remodeling and the potential roles of different autonomic modulation strategies on atrial metabolic aberrations in AF.


Subject(s)
Atrial Fibrillation , Humans , Autonomic Nervous System , Heart Atria , Heart Rate/physiology
20.
J Thorac Cardiovasc Surg ; 165(4): e158-e174, 2023 04.
Article in English | MEDLINE | ID: mdl-35461705

ABSTRACT

OBJECTIVES: The mechanisms underlying atrial fibrillation are yet to be elucidated. We sought to investigate the interactions among autonomic remodeling, epicardial adipose tissue, inflammation, and atrial fibrillation. METHODS: Myocardium and adjacent epicardial adipose tissue of the left atrial appendage, right atrial appendage, and pulmonary vein muscle sleeves were obtained from 61 consecutive patients (35 with atrial fibrillation, 26 with no atrial fibrillation) during mitral valve surgeries. Patients were divided into the atrial fibrillation group and no atrial fibrillation group according to the history and Holter monitoring before surgery. Sympathetic and parasympathetic innervation were evaluated by tyrosine hydroxylase and choline acetyltransferase staining, respectively. Atrial fibrosis as well as cytokines/adipokines and related inflammatory proteins and signaling pathways in the epicardial adipose tissue were examined. RESULTS: Immunohistochemical studies revealed significantly increased tyrosine hydroxylase (+) and choline acetyltransferase (+) neural elements in the left atrial appendage and pulmonary vein muscle sleeve myocardium, as well as adjacent epicardial adipose tissue in the atrial fibrillation group, particularly the pulmonary vein muscle sleeve sites. The receiver operating curve identified a threshold ratio (tyrosine hydroxylase/choline acetyltransferase) of 0.8986 in the epicardial adipose tissue (sensitivity = 82.86%; specificity = 80.77%; area under the curve = 0.85, 95% confidence interval = 0.76-0.95, P < .0001). More patients with a higher tyrosine hydroxylase/choline acetyltransferase ratio (≥0.8986) had atrial fibrillation. Expression levels of the genes and related proteins of the ß1 adrenergic, mitogen-activated protein kinase, and nuclear factor kappa B signaling pathways were higher in patients with a higher tyrosine hydroxylase/choline acetyltransferase ratio. The tyrosine hydroxylase/choline acetyltransferase ratio also correlated with fibrosis. CONCLUSIONS: Differentially enhanced autonomic remodeling and proinflammatory and profibrotic cytokines/adipokines in the epicardial adipose tissue adjacent to the pulmonary vein muscle sleeve site may work synergistically to promote atrial fibrillation.


Subject(s)
Atrial Fibrillation , Tyrosine 3-Monooxygenase , Humans , Tyrosine 3-Monooxygenase/metabolism , Choline O-Acetyltransferase/metabolism , Atrial Fibrillation/surgery , Heart Atria , Pericardium/metabolism , Cytokines/metabolism , Fibrosis , Adipokines/metabolism , Adipose Tissue
SELECTION OF CITATIONS
SEARCH DETAIL