Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
1.
J Cardiovasc Electrophysiol ; 34(8): 1613-1621, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37365931

RESUMO

INTRODUCTION: Improved sinus rhythm (SR) maintenance rates have been achieved in patients with persistent atrial fibrillation (AF) undergoing pulmonary vein isolation plus additional ablation of low voltage substrate (LVS) during SR. However, voltage mapping during SR may be hindered in persistent and long-persistent AF patients by immediate AF recurrence after electrical cardioversion. We assess correlations between LVS extent and location during SR and AF, aiming to identify regional voltage thresholds for rhythm-independent delineation/detection of LVS areas. (1) Identification of voltage dissimilarities between mapping in SR and AF. (2) Identification of regional voltage thresholds that improve cross-rhythm substrate detection. (3) Comparison of LVS between SR and native versus induced AF. METHODS: Forty-one ablation-naive persistent AF patients underwent high-definition (1 mm electrodes; >1200 left atrial (LA) mapping sites per rhythm) voltage mapping in SR and AF. Global and regional voltage thresholds in AF were identified which best match LVS < 0.5 mV and <1.0 mV in SR. Additionally, the correlation between SR-LVS with induced versus native AF-LVS was assessed. RESULTS: Substantial voltage differences (median: 0.52, interquartile range: 0.33-0.69, maximum: 1.19 mV) with a predominance of the posterior/inferior LA wall exist between the rhythms. An AF threshold of 0.34 mV for the entire left atrium provides an accuracy, sensitivity and specificity of 69%, 67%, and 69% to identify SR-LVS < 0.5 mV, respectively. Lower thresholds for the posterior wall (0.27 mV) and inferior wall (0.3 mV) result in higher spatial concordance to SR-LVS (4% and 7% increase). Concordance with SR-LVS was higher for induced AF compared to native AF (area under the curve[AUC]: 0.80 vs. 0.73). AF-LVS < 0.5 mV corresponds to SR-LVS < 0.97 mV (AUC: 0.73). CONCLUSION: Although the proposed region-specific voltage thresholds during AF improve the consistency of LVS identification as determined during SR, the concordance in LVS between SR and AF remains moderate, with larger LVS detection during AF. Voltage-based substrate ablation should preferentially be performed during SR to limit the amount of ablated atrial myocardium.


Assuntos
Apêndice Atrial , Fibrilação Atrial , Ablação por Cateter , Humanos , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/cirurgia , Técnicas Eletrofisiológicas Cardíacas , Átrios do Coração/cirurgia
2.
Europace ; 25(1): 211-222, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35943361

RESUMO

AIMS: The long-term success rate of ablation therapy is still sub-optimal in patients with persistent atrial fibrillation (AF), mostly due to arrhythmia recurrence originating from arrhythmogenic sites outside the pulmonary veins. Computational modelling provides a framework to integrate and augment clinical data, potentially enabling the patient-specific identification of AF mechanisms and of the optimal ablation sites. We developed a technology to tailor ablations in anatomical and functional digital atrial twins of patients with persistent AF aiming to identify the most successful ablation strategy. METHODS AND RESULTS: Twenty-nine patient-specific computational models integrating clinical information from tomographic imaging and electro-anatomical activation time and voltage maps were generated. Areas sustaining AF were identified by a personalized induction protocol at multiple locations. State-of-the-art anatomical and substrate ablation strategies were compared with our proposed Personalized Ablation Lines (PersonAL) plan, which consists of iteratively targeting emergent high dominant frequency (HDF) regions, to identify the optimal ablation strategy. Localized ablations were connected to the closest non-conductive barrier to prevent recurrence of AF or atrial tachycardia. The first application of the HDF strategy had a success of >98% and isolated only 5-6% of the left atrial myocardium. In contrast, conventional ablation strategies targeting anatomical or structural substrate resulted in isolation of up to 20% of left atrial myocardium. After a second iteration of the HDF strategy, no further arrhythmia episode could be induced in any of the patient-specific models. CONCLUSION: The novel PersonAL in silico technology allows to unveil all AF-perpetuating areas and personalize ablation by leveraging atrial digital twins.


Assuntos
Fibrilação Atrial , Ablação por Cateter , Veias Pulmonares , Taquicardia Supraventricular , Humanos , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/cirurgia , Resultado do Tratamento , Átrios do Coração/cirurgia , Simulação por Computador , Veias Pulmonares/cirurgia , Ablação por Cateter/efeitos adversos , Ablação por Cateter/métodos , Recidiva
3.
Europace ; 25(9)2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37713626

RESUMO

AIMS: Electro-anatomical voltage, conduction velocity (CV) mapping, and late gadolinium enhancement (LGE) magnetic resonance imaging (MRI) have been correlated with atrial cardiomyopathy (ACM). However, the comparability between these modalities remains unclear. This study aims to (i) compare pathological substrate extent and location between current modalities, (ii) establish spatial histograms in a cohort, (iii) develop a new estimated optimized image intensity threshold (EOIIT) for LGE-MRI identifying patients with ACM, (iv) predict rhythm outcome after pulmonary vein isolation (PVI) for persistent atrial fibrillation (AF). METHODS AND RESULTS: Thirty-six ablation-naive persistent AF patients underwent LGE-MRI and high-definition electro-anatomical mapping in sinus rhythm. Late gadolinium enhancement areas were classified using the UTAH, image intensity ratio (IIR >1.20), and new EOIIT method for comparison to low-voltage substrate (LVS) and slow conduction areas <0.2 m/s. Receiver operating characteristic analysis was used to determine LGE thresholds optimally matching LVS. Atrial cardiomyopathy was defined as LVS extent ≥5% of the left atrium (LA) surface at <0.5 mV. The degree and distribution of detected pathological substrate (percentage of individual LA surface are) varied significantly (P < 0.001) across the mapping modalities: 10% (interquartile range 0-14%) of the LA displayed LVS <0.5 mV vs. 7% (0-12%) slow conduction areas <0.2 m/s vs. 15% (8-23%) LGE with the UTAH method vs. 13% (2-23%) using IIR >1.20, with most discrepancies on the posterior LA. Optimized image intensity thresholds and each patient's mean blood pool intensity correlated linearly (R2 = 0.89, P < 0.001). Concordance between LGE-MRI-based and LVS-based ACM diagnosis improved with the novel EOIIT applied at the anterior LA [83% sensitivity, 79% specificity, area under the curve (AUC): 0.89] in comparison to the UTAH method (67% sensitivity, 75% specificity, AUC: 0.81) and IIR >1.20 (75% sensitivity, 62% specificity, AUC: 0.67). CONCLUSION: Discordances in detected pathological substrate exist between LVS, CV, and LGE-MRI in the LA, irrespective of the LGE detection method. The new EOIIT method improves concordance of LGE-MRI-based ACM diagnosis with LVS in ablation-naive AF patients but discrepancy remains particularly on the posterior wall. All methods may enable the prediction of rhythm outcomes after PVI in patients with persistent AF.


Assuntos
Fibrilação Atrial , Cardiomiopatias , Ablação por Cateter , Humanos , Fibrilação Atrial/diagnóstico por imagem , Fibrilação Atrial/cirurgia , Meios de Contraste , Gadolínio , Estudos de Coortes , Átrios do Coração/diagnóstico por imagem , Átrios do Coração/cirurgia , Átrios do Coração/patologia , Imageamento por Ressonância Magnética/métodos , Cardiomiopatias/etiologia , Ablação por Cateter/efeitos adversos
4.
Europace ; 24(7): 1186-1194, 2022 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-35045172

RESUMO

AIMS: Atrial flutter (AFlut) is a common re-entrant atrial tachycardia driven by self-sustainable mechanisms that cause excitations to propagate along pathways different from sinus rhythm. Intra-cardiac electrophysiological mapping and catheter ablation are often performed without detailed prior knowledge of the mechanism perpetuating AFlut, likely prolonging the procedure time of these invasive interventions. We sought to discriminate the AFlut location [cavotricuspid isthmus-dependent (CTI), peri-mitral, and other left atrium (LA) AFlut classes] with a machine learning-based algorithm using only the non-invasive signals from the 12-lead electrocardiogram (ECG). METHODS AND RESULTS: Hybrid 12-lead ECG dataset of 1769 signals was used (1424 in silico ECGs, and 345 clinical ECGs from 115 patients-three different ECG segments over time were extracted from each patient corresponding to single AFlut cycles). Seventy-seven features were extracted. A decision tree classifier with a hold-out classification approach was trained, validated, and tested on the dataset randomly split after selecting the most informative features. The clinical test set comprised 38 patients (114 clinical ECGs). The classifier yielded 76.3% accuracy on the clinical test set with a sensitivity of 89.7%, 75.0%, and 64.1% and a positive predictive value of 71.4%, 75.0%, and 86.2% for CTI, peri-mitral, and other LA class, respectively. Considering majority vote of the three segments taken from each patient, the CTI class was correctly classified at 92%. CONCLUSION: Our results show that a machine learning classifier relying only on non-invasive signals can potentially identify the location of AFlut mechanisms. This method could aid in planning and tailoring patient-specific AFlut treatments.


Assuntos
Flutter Atrial , Ablação por Cateter , Flutter Atrial/diagnóstico , Flutter Atrial/etiologia , Flutter Atrial/cirurgia , Eletrocardiografia/métodos , Sistema de Condução Cardíaco , Humanos , Aprendizado de Máquina
5.
Europace ; 23(23 Suppl 1): i133-i142, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33751084

RESUMO

AIMS: The treatment of atrial fibrillation beyond pulmonary vein isolation has remained an unsolved challenge. Targeting regions identified by different substrate mapping approaches for ablation resulted in ambiguous outcomes. With the effective refractory period being a fundamental prerequisite for the maintenance of fibrillatory conduction, this study aims at estimating the effective refractory period with clinically available measurements. METHODS AND RESULTS: A set of 240 simulations in a spherical model of the left atrium with varying model initialization, combination of cellular refractory properties, and size of a region of lowered effective refractory period was implemented to analyse the capabilities and limitations of cycle length mapping. The minimum observed cycle length and the 25% quantile were compared to the underlying effective refractory period. The density of phase singularities was used as a measure for the complexity of the excitation pattern. Finally, we employed the method in a clinical test of concept including five patients. Areas of lowered effective refractory period could be distinguished from their surroundings in simulated scenarios with successfully induced multi-wavelet re-entry. Larger areas and higher gradients in effective refractory period as well as complex activation patterns favour the method. The 25% quantile of cycle lengths in patients with persistent atrial fibrillation was found to range from 85 to 190 ms. CONCLUSION: Cycle length mapping is capable of highlighting regions of pathologic refractory properties. In combination with complementary substrate mapping approaches, the method fosters confidence to enhance the treatment of atrial fibrillation beyond pulmonary vein isolation particularly in patients with complex activation patterns.


Assuntos
Fibrilação Atrial , Ablação por Cateter , Veias Pulmonares , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/cirurgia , Simulação por Computador , Átrios do Coração , Frequência Cardíaca , Humanos , Veias Pulmonares/cirurgia
6.
Biomed Eng Online ; 20(1): 69, 2021 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294108

RESUMO

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is typically caused by mutations in sarcomeric genes leading to cardiomyocyte disarray, replacement fibrosis, impaired contractility, and elevated filling pressures. These varying tissue properties are associated with certain strain patterns that may allow to establish a diagnosis by means of non-invasive imaging without the necessity of harmful myocardial biopsies or contrast agent application. With a numerical study, we aim to answer: how the variability in each of these mechanisms contributes to altered mechanics of the left ventricle (LV) and if the deformation obtained in in-silico experiments is comparable to values reported from clinical measurements. METHODS: We conducted an in-silico sensitivity study on physiological and pathological mechanisms potentially underlying the clinical HCM phenotype. The deformation of the four-chamber heart models was simulated using a finite-element mechanical solver with a sliding boundary condition to mimic the tissue surrounding the heart. Furthermore, a closed-loop circulatory model delivered the pressure values acting on the endocardium. Deformation measures and mechanical behavior of the heart models were evaluated globally and regionally. RESULTS: Hypertrophy of the LV affected the course of strain, strain rate, and wall thickening-the root-mean-squared difference of the wall thickening between control (mean thickness 10 mm) and hypertrophic geometries (17 mm) was >10%. A reduction of active force development by 40% led to less overall deformation: maximal radial strain reduced from 26 to 21%. A fivefold increase in tissue stiffness caused a more homogeneous distribution of the strain values among 17 heart segments. Fiber disarray led to minor changes in the circumferential and radial strain. A combination of pathological mechanisms led to reduced and slower deformation of the LV and halved the longitudinal shortening of the LA. CONCLUSIONS: This study uses a computer model to determine the changes in LV deformation caused by pathological mechanisms that are presumed to underlay HCM. This knowledge can complement imaging-derived information to obtain a more accurate diagnosis of HCM.


Assuntos
Cardiomiopatia Hipertrófica , Ventrículos do Coração , Cardiomiopatia Hipertrófica/diagnóstico por imagem , Meios de Contraste , Coração , Ventrículos do Coração/diagnóstico por imagem , Humanos
7.
J Therm Biol ; 84: 316-322, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31466769

RESUMO

Local brain hypothermia is an attractive method for providing cerebral neuroprotection for ischemic stroke patients and at the same time reducing systemic side effects of cooling. In acute ischemic stroke patients with large vessel occlusion, combination with endovascular mechanical recanalization treatment could potentially allow for an alleviation of inflammatory and apoptotic pathways in the critical phase of reperfusion. The direct cooling of arterial blood by means of an intra-carotid heat exchange catheter compatible with recanalization systems is a novel promising approach. Focusing on the concept of "cold reperfusion", we developed an energetic model to calculate the rate of temperature decrease during intra-carotid cooling in case of physiological as well as decreased perfusion. Additionally, we discussed and considered the effect and biological significance of temperature decrease on resulting brain perfusion. Our model predicted a 2 °C brain temperature decrease in 8.3, 11.8 and 26.2 min at perfusion rates of 50, 30 and 10ml100g⋅min, respectively. The systemic temperature decrease - caused by the venous blood return to the main circulation - was limited to 0.5 °C in 60 min. Our results underline the potential of catheter-assisted, intracarotid blood cooling to provide a fast and selective brain temperature decrease in the phase of vessel recanalization. This method can potentially allow for a tissue hypothermia during the restoration of the physiological flow and thus a "cold reperfusion" in the setting of mechanical recanalization.


Assuntos
Temperatura Corporal , Encéfalo/irrigação sanguínea , Encéfalo/fisiologia , Hipotermia Induzida , Infarto da Artéria Cerebral Média/terapia , Modelos Biológicos , Acidente Vascular Cerebral/terapia , Animais , Cateterismo , Humanos
8.
Europace ; 20(suppl_3): iii36-iii44, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30476059

RESUMO

AIMS: Chronic left atrial enlargement (LAE) increases the risk of atrial fibrillation. Electrocardiogram (ECG) criteria might provide a means to diagnose LAE and identify patients at risk; however, current criteria perform poorly. We seek to characterize the potentially differential effects of atrial dilation vs. hypertrophy on the ECG P-wave. METHODS AND RESULTS: We predict effects on the P-wave of (i) left atrial dilation (LAD), i.e. an increase of LA cavity volume without an increase in myocardial volume, (ii) left atrial concentric hypertrophy (LACH), i.e. a thickened myocardial wall, and (iii) a combination of the two. We performed a computational study in a cohort of 72 anatomical variants, derived from four human atrial anatomies. To model LAD, pressure was applied to the LA endocardium increasing cavity volume by up to 100%. For LACH, the LA wall was thickened by up to 3.3 mm. P-waves were derived by simulating atrial excitation propagation and computing the body surface ECG. The sensitivity regarding changes beyond purely anatomical effects was analysed by altering conduction velocity by 25% in 96 additional model variants. Left atrial dilation prolonged P-wave duration (PWd) in two of four subjects; in one subject a shortening, and in the other a variable change were seen. Left atrial concentric hypertrophy, in contrast, consistently increased P-wave terminal force in lead V1 (PTF-V1) in all subjects through an enlarged amplitude while PWd was unaffected. Combined hypertrophy and dilation generally enhanced the effect of hypertrophy on PTF-V1. CONCLUSION: Isolated LAD has moderate effects on the currently used P-wave criteria, explaining the limited utility of PWd and PTF-V1 in detecting LAE in clinical practice. In contrast, PTF-V1 may be a more sensitive indicator of LA myocardial hypertrophy.


Assuntos
Potenciais de Ação , Fibrilação Atrial/diagnóstico , Função do Átrio Esquerdo , Remodelamento Atrial , Cardiomegalia/diagnóstico , Eletrocardiografia , Átrios do Coração/fisiopatologia , Frequência Cardíaca , Fibrilação Atrial/etiologia , Fibrilação Atrial/fisiopatologia , Cardiomegalia/complicações , Cardiomegalia/fisiopatologia , Simulação por Computador , Diagnóstico Diferencial , Átrios do Coração/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Modelos Cardiovasculares , Valor Preditivo dos Testes , Fatores de Risco , Fatores de Tempo
9.
Europace ; 18(suppl 4): iv35-iv43, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28011829

RESUMO

AIMS: P-wave morphology correlates with the risk for atrial fibrillation (AF). Left atrial (LA) enlargement could explain both the higher risk for AF and higher P-wave terminal force (PTF) in lead V1. However, PTF-V1 has been shown to correlate poorly with LA size. We hypothesize that PTF-V1 is also affected by the earliest activated site (EAS) in the right atrium and its proximity to inter-atrial connections (IAC), which both show tremendous variability. METHODS AND RESULTS: Atrial excitation was triggered from seven different EAS in a cohort of eight anatomically personalized computational models. The posterior IACs were non-conductive in a second set of simulations. Body surface ECGs were computed and separated by left and right atrial contributions. Mid-septal EAS yielded the highest PTF-V1. More anterior/superior and more inferior EAS yielded lower absolute PTF-V1 values deviating by a factor of up to 2.0 for adjacent EAS. Earliest right-to-left activation was conducted via Bachmann's Bundle (BB) for anterior/superior EAS and shifted towards posterior IACs for more inferior EAS. Non-conducting posterior IACs increased PTF-V1 by up to 150% compared to intact posterior IACs for inferior EAS. LA contribution to the P-wave integral was 24% on average. CONCLUSION: The electrical contributor's site of earliest activation and intactness of posterior IACs affect PTF-V1 significantly by changing LA breakthrough sites independent from LA size. This should be considered for interpretation of electrocardiographical signs of LA abnormality and LA enlargement.


Assuntos
Potenciais de Ação , Fibrilação Atrial/fisiopatologia , Função do Átrio Direito , Átrios do Coração/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Modelos Cardiovasculares , Modelagem Computacional Específica para o Paciente , Adulto , Idoso , Fibrilação Atrial/diagnóstico , Função do Átrio Esquerdo , Eletrocardiografia , Frequência Cardíaca , Humanos , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Processamento de Sinais Assistido por Computador , Fatores de Tempo , Adulto Jovem
10.
J Magn Reson Imaging ; 42(4): 964-71, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25639861

RESUMO

PURPOSE: To evaluate two commonly used respiratory motion correction techniques for coronary magnetic resonance angiography (MRA) regarding their dependency on motion estimation accuracy and final image quality and to compare both methods to the respiratory gating approach used in clinical practice. MATERIALS AND METHODS: Ten healthy volunteers were scanned using a non-Cartesian radial phase encoding acquisition. Respiratory motion was corrected for coronary MRA according to two motion correction techniques, image-based (IMC) and reconstruction-based (RMC) respiratory motion correction. Both motion correction approaches were compared quantitatively and qualitatively against a reference standard navigator-based respiratory gating (RG) approach. Quantitative comparisons were performed regarding visible vessel length, vessel sharpness, and total acquisition time. Two experts carried out a visual scoring of image quality. Additionally, numerical simulations were performed to evaluate the effect of motion estimation inaccuracy on RMC and IMC. RESULTS: RMC led to significantly better image quality than IMC (P's paired Student's t-test were smaller than 0.001 for vessel sharpness and visual scoring). RMC did not show a statistically significant difference compared to reference standard RG (vessel length [99% confidence interval]: 86.913 [83.097-95.015], P = 0.107; vessel sharpness: 0.640 [0.605-0.802], P = 0.012; visual scoring: 2.583 [2.410-3.424], P = 0.018) in terms of vessel visualization and image quality while reducing scan times by 56%. Simulations showed higher dependencies for RMC than for IMC on motion estimation inaccuracies. CONCLUSION: RMC provides a similar image quality as the clinically used RG approach but almost halves the scan time and is independent of subjects' breathing patterns. Clinical validation of RMC is now desirable.


Assuntos
Artefatos , Angiografia Coronária/métodos , Vasos Coronários/anatomia & histologia , Aumento da Imagem/métodos , Angiografia por Ressonância Magnética/métodos , Técnicas de Imagem de Sincronização Respiratória/métodos , Adulto , Algoritmos , Feminino , Humanos , Interpretação de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Armazenamento e Recuperação da Informação/métodos , Masculino , Movimento (Física) , Variações Dependentes do Observador , Reprodutibilidade dos Testes , Mecânica Respiratória , Sensibilidade e Especificidade , Processamento de Sinais Assistido por Computador
11.
J Electrocardiol ; 48(2): 171-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25595719

RESUMO

BACKGROUND: Intracardiac electrograms are an indispensable part during diagnosis of supraventricular arrhythmias, but atrial activity (AA) can be obscured by ventricular far-fields (VFF). Concepts based on statistical independence like principal component analysis (PCA) cannot be applied for VFF removal during atrial tachycardia with stable conduction. METHODS: A database of realistic electrograms containing AA and VFF was generated. Both PCA and the new technique periodic component analysis (πCA) were implemented, benchmarked, and applied to clinical data. RESULTS: The concept of πCA was successfully verified to retain compromised AA morphology, showing high correlation (cc=0.98±0.01) for stable atrial cycle length (ACL). Performance of PCA failed during temporal coupling (cc=0.03±0.08) but improved for increasing conduction variability (cc=0.77±0.14). Stability of ACL was identified as a critical parameter for πCA application. Analysis of clinical data confirmed these findings. CONCLUSION: πCA is introduced as a powerful new technique for artifact removal in periodic signals. Its concept and performance were benchmarked against PCA using simulated data and demonstrated on measured electrograms.


Assuntos
Artefatos , Flutter Atrial/diagnóstico , Eletrocardiografia/métodos , Idoso , Flutter Atrial/fisiopatologia , Benchmarking , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Componente Principal , Processamento de Sinais Assistido por Computador , Taquicardia Supraventricular/diagnóstico , Taquicardia Supraventricular/fisiopatologia
12.
Europace ; 16(3): 435-43, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24569898

RESUMO

AIMS: Human ether-à-go-go-related gene (hERG) missense mutations N588K and L532P are both associated with atrial fibrillation (AF). However, the underlying gain-of-function mechanism is different. The aim of this computational study is to assess and understand the arrhythmogenic mechanisms of these genetic disorders on the cellular and tissue level as a basis for the improvement of therapeutic strategies. METHODS AND RESULTS: The IKr formulation of an established model of human atrial myocytes was adapted by using the measurement data of wild-type and mutant hERG channels. Restitution curves of the action potential duration and its slope, effective refractory period (ERP), conduction velocity, reentry wavelength (WL), and the vulnerable window (VW) were determined in a one-dimensional (1D) tissue strand. Moreover, spiral wave inducibility and rotor lifetime in a 2D tissue patch were evaluated. The two mutations caused an increase in IKr regarding both peak amplitude and current integral, whereas the duration during which IKr is active was decreased. The WL was reduced due to a shorter ERP. Spiral waves could be initiated by using mutation models as opposed to the control case. The frequency dependency of the VW was reversed. CONCLUSION: Both mutations showed an increased arrhythmogenicity due to decreased refractory time in combination with a more linear repolarization phase. The effects were more pronounced for mutation L532P than for N588K. Furthermore, spiral waves presented higher stability and a more regular pattern for L532P. These in silico investigations unveiling differences of mutations affecting the same ion channel may help to advance genotype-guided AF prevention and therapy strategies.


Assuntos
Fibrilação Atrial/fisiopatologia , Canais de Potássio Éter-A-Go-Go/genética , Átrios do Coração/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Modelos Cardiovasculares , Modelos Genéticos , Células Musculares , Potenciais de Ação/genética , Simulação por Computador , Canal de Potássio ERG1 , Humanos , Ativação do Canal Iônico/genética , Potenciais da Membrana/genética , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética
13.
Europace ; 16 Suppl 4: iv30-iv38, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25362168

RESUMO

AIMS: The clinical efficacy in preventing the recurrence of atrial fibrillation (AF) is higher for amiodarone than for dronedarone. Moreover, pharmacotherapy with these drugs is less successful in patients with remodelled substrate induced by chronic AF (cAF) and patients suffering from familial AF. To date, the reasons for these phenomena are only incompletely understood. We analyse the effects of the drugs in a computational model of atrial electrophysiology. METHODS AND RESULTS: The Courtemanche-Ramirez-Nattel model was adapted to represent cAF remodelled tissue and hERG mutations N588K and L532P. The pharmacodynamics of amiodarone and dronedarone were investigated with respect to their dose and heart rate dependence by evaluating 10 descriptors of action potential morphology and conduction properties. An arrhythmia score was computed based on a subset of these biomarkers and analysed regarding circadian variation of drug concentration and heart rate. Action potential alternans at high frequencies was observed over the whole dronedarone concentration range at high frequencies, while amiodarone caused alternans only in a narrow range. The total score of dronedarone reached critical values in most of the investigated dynamic scenarios, while amiodarone caused only minor score oscillations. Compared with the other substrates, cAF showed significantly different characteristics resulting in a lower amiodarone but higher dronedarone concentration yielding the lowest score. CONCLUSION: Significant differences exist in the frequency and concentration-dependent effects between amiodarone and dronedarone and between different atrial substrates. Our results provide possible explanations for the superior efficacy of amiodarone and may aid in the design of substrate-specific pharmacotherapy for AF.


Assuntos
Amiodarona/análogos & derivados , Amiodarona/uso terapêutico , Antiarrítmicos/uso terapêutico , Fibrilação Atrial/terapia , Simulação por Computador , Átrios do Coração/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Modelos Cardiovasculares , Potenciais de Ação , Amiodarona/farmacocinética , Antiarrítmicos/farmacocinética , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/genética , Fibrilação Atrial/metabolismo , Fibrilação Atrial/fisiopatologia , Relação Dose-Resposta a Droga , Dronedarona , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Átrios do Coração/fisiopatologia , Humanos , Mutação , Análise Numérica Assistida por Computador , Recidiva , Fatores de Tempo , Resultado do Tratamento
14.
Artif Intell Med ; 143: 102619, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37673581

RESUMO

Cardiovascular diseases account for 17 million deaths per year worldwide. Of these, 25% are categorized as sudden cardiac death, which can be related to ventricular tachycardia (VT). This type of arrhythmia can be caused by focal activation sources outside the sinus node. Catheter ablation of these foci is a curative treatment in order to inactivate the abnormal triggering activity. However, the localization procedure is usually time-consuming and requires an invasive procedure in the catheter lab. To facilitate and expedite the treatment, we present two novel localization support techniques based on convolutional neural networks (CNNs) that address these clinical needs. In contrast to existing methods, our approaches were designed to be independent of the patient-specific geometry and directly applicable to surface ECG signals, while also delivering a binary transmural position. Moreover, one of the method's outputs can be interpreted as several ranked solutions. The CNNs were trained on a dataset containing only simulated data and evaluated both on simulated test data and clinical data. On a novel large and open simulated dataset, the median test error was below 3 mm. The median localization error on the unseen clinical data ranged from 32 mm to 41 mm without optimizing the pre-processing and CNN to the clinical data. Interpreting the output of one of the approaches as ranked solutions, the best median error of the top-3 solutions decreased to 20 mm on the clinical data. The transmural position was correctly detected in up to 82% of all clinical cases. These results demonstrate a proof of principle to utilize CNNs to localize the activation source without the intrinsic need for patient-specific geometrical information. Furthermore, providing multiple solutions can assist physicians in identifying the true activation source amongst more than one possible location. With further optimization to clinical data, these methods have high potential to accelerate clinical interventions, replace certain steps within these procedures and consequently reduce procedural risk and improve VT patient outcomes.


Assuntos
Aprendizado Profundo , Médicos , Humanos , Redes Neurais de Computação , Pacientes
15.
IEEE Trans Biomed Eng ; 70(2): 511-522, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35921339

RESUMO

OBJECTIVE: The bidomain model and the finite element method are an established standard to mathematically describe cardiac electrophysiology, but are both suboptimal choices for fast and large-scale simulations due to high computational costs. We investigate to what extent simplified approaches for propagation models (monodomain, reaction-Eikonal and Eikonal) and forward calculation (boundary element and infinite volume conductor) deliver markedly accelerated, yet physiologically accurate simulation results in atrial electrophysiology. METHODS: We compared action potential durations, local activation times (LATs), and electrocardiograms (ECGs) for sinus rhythm simulations on healthy and fibrotically infiltrated atrial models. RESULTS: All simplified model solutions yielded LATs and P waves in accurate accordance with the bidomain results. Only for the Eikonal model with pre-computed action potential templates shifted in time to derive transmembrane voltages, repolarization behavior notably deviated from the bidomain results. ECGs calculated with the boundary element method were characterized by correlation coefficients 0.9 compared to the finite element method. The infinite volume conductor method led to lower correlation coefficients caused predominantly by systematic overestimations of P wave amplitudes in the precordial leads. CONCLUSION: Our results demonstrate that the Eikonal model yields accurate LATs and combined with the boundary element method precise ECGs compared to markedly more expensive full bidomain simulations. However, for an accurate representation of atrial repolarization dynamics, diffusion terms must be accounted for in simplified models. SIGNIFICANCE: Simulations of atrial LATs and ECGs can be notably accelerated to clinically feasible time frames at high accuracy by resorting to the Eikonal and boundary element methods.


Assuntos
Fibrilação Atrial , Sistema de Condução Cardíaco , Humanos , Sistema de Condução Cardíaco/fisiologia , Modelos Cardiovasculares , Átrios do Coração , Simulação por Computador , Eletrofisiologia Cardíaca , Coração/fisiologia
16.
IEEE Trans Biomed Eng ; 70(2): 533-543, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35925848

RESUMO

BACKGROUND: Electrical impedance measurements have become an accepted tool for monitoring intracardiac radio frequency ablation. Recently, the long-established generator impedance was joined by novel local impedance measurement capabilities with all electrical circuit terminals being accommodated within the catheter. OBJECTIVE: This work aims at in silico quantification of distinct influencing factors that have remained challenges due to the lack of ground truth knowledge and the superposition of effects in clinical settings. METHODS: We introduced a highly detailed in silico model of two local impedance enabled catheters, namely IntellaNav MiFi OI and IntellaNav Stablepoint, embedded in a series of clinically relevant environments. Assigning material and frequency specific conductivities and subsequently calculating the spread of the electrical field with the finite element method yielded in silico local impedances. The in silico model was validated by comparison to in vitro measurements of standardized sodium chloride solutions. We then investigated the effect of the withdrawal of the catheter into the transseptal sheath, catheter-tissue interaction, insertion of the catheter into pulmonary veins, and catheter irrigation. RESULTS: All simulated setups were in line with in vitro experiments and in human measurements and gave detailed insight into determinants of local impedance changes as well as the relation between values measured with two different devices. CONCLUSION: The in silico environment proved to be capable of resembling clinical scenarios and quantifying local impedance changes. SIGNIFICANCE: The tool can assists the interpretation of measurements in humans and has the potential to support future catheter development.


Assuntos
Ablação por Cateter , Átrios do Coração , Humanos , Impedância Elétrica , Condutividade Elétrica , Catéteres , Simulação por Computador , Ablação por Cateter/métodos
17.
Comput Med Imaging Graph ; 108: 102265, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37392493

RESUMO

Digital twins of patients' hearts are a promising tool to assess arrhythmia vulnerability and to personalize therapy. However, the process of building personalized computational models can be challenging and requires a high level of human interaction. We propose a patient-specific Augmented Atria generation pipeline (AugmentA) as a highly automated framework which, starting from clinical geometrical data, provides ready-to-use atrial personalized computational models. AugmentA identifies and labels atrial orifices using only one reference point per atrium. If the user chooses to fit a statistical shape model to the input geometry, it is first rigidly aligned with the given mean shape before a non-rigid fitting procedure is applied. AugmentA automatically generates the fiber orientation and finds local conduction velocities by minimizing the error between the simulated and clinical local activation time (LAT) map. The pipeline was tested on a cohort of 29 patients on both segmented magnetic resonance images (MRI) and electroanatomical maps of the left atrium. Moreover, the pipeline was applied to a bi-atrial volumetric mesh derived from MRI. The pipeline robustly integrated fiber orientation and anatomical region annotations in 38.4 ± 5.7 s. In conclusion, AugmentA offers an automated and comprehensive pipeline delivering atrial digital twins from clinical data in procedural time.


Assuntos
Fibrilação Atrial , Humanos , Átrios do Coração/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos
18.
Sci Data ; 10(1): 279, 2023 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-37179420

RESUMO

Machine learning (ML) methods for the analysis of electrocardiography (ECG) data are gaining importance, substantially supported by the release of large public datasets. However, these current datasets miss important derived descriptors such as ECG features that have been devised in the past hundred years and still form the basis of most automatic ECG analysis algorithms and are critical for cardiologists' decision processes. ECG features are available from sophisticated commercial software but are not accessible to the general public. To alleviate this issue, we add ECG features from two leading commercial algorithms and an open-source implementation supplemented by a set of automatic diagnostic statements from a commercial ECG analysis software in preprocessed format. This allows the comparison of ML models trained on clinically versus automatically generated label sets. We provide an extensive technical validation of features and diagnostic statements for ML applications. We believe this release crucially enhances the usability of the PTB-XL dataset as a reference dataset for ML methods in the context of ECG data.


Assuntos
Algoritmos , Eletrocardiografia , Software , Eletrocardiografia/métodos , Aprendizado de Máquina , Humanos
19.
Sci Data ; 10(1): 531, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37553349

RESUMO

Mechanistic cardiac electrophysiology models allow for personalized simulations of the electrical activity in the heart and the ensuing electrocardiogram (ECG) on the body surface. As such, synthetic signals possess known ground truth labels of the underlying disease and can be employed for validation of machine learning ECG analysis tools in addition to clinical signals. Recently, synthetic ECGs were used to enrich sparse clinical data or even replace them completely during training leading to improved performance on real-world clinical test data. We thus generated a novel synthetic database comprising a total of 16,900 12 lead ECGs based on electrophysiological simulations equally distributed into healthy control and 7 pathology classes. The pathological case of myocardial infraction had 6 sub-classes. A comparison of extracted features between the virtual cohort and a publicly available clinical ECG database demonstrated that the synthetic signals represent clinical ECGs for healthy and pathological subpopulations with high fidelity. The ECG database is split into training, validation, and test folds for development and objective assessment of novel machine learning algorithms.


Assuntos
Eletrocardiografia , Coração , Humanos , Algoritmos , Aprendizado de Máquina , Miocárdio
20.
Europace ; 14 Suppl 5: v90-v96, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23104920

RESUMO

AIMS: Amiodarone and cisapride are both known to prolong the QT interval, yet the two drugs have different effects on arrhythmia. Cisapride can cause torsades de pointes while amiodarone is found to be anti-arrhythmic. A computational model was used to investigate the action of these two drugs. METHODS AND RESULTS: In a biophysically detailed model, the ion current conductivities affected by both drugs were reduced in order to simulate the pharmacological effects in healthy and ischaemic cells. Furthermore, restitution curves of the action potential duration (APD), effective refractory period, conduction velocity, wavelength, and the vulnerable window were determined in a one-dimensional (1D) tissue strand. Moreover, cardiac excitation propagation was computed in a 3D model of healthy ventricles. The corresponding body surface potentials were calculated and standard 12-lead electrocardiograms were derived. Both cisapride and amiodarone caused a prolongation of the QT interval and the refractory period. However, cisapride did not significantly alter the conduction-related properties, such as e.g. the wavelength or vulnerable window, whereas amiodarone had a larger impact on them. It slightly flattened the APD restitution slope and furthermore reduced the conduction velocity and wavelength. CONCLUSION: Both drugs show similar prolongation of the QT interval, although they present different electrophysiological properties in the single-cell as well as in tissue simulations of cardiac excitation propagation. These computer simulations help to better understand the underlying mechanisms responsible for the initiation or termination of arrhythmias caused by amiodarone and cisapride.


Assuntos
Amiodarona/administração & dosagem , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/fisiopatologia , Cisaprida/administração & dosagem , Sistema de Condução Cardíaco/fisiopatologia , Ventrículos do Coração/fisiopatologia , Modelos Cardiovasculares , Antiarrítmicos/administração & dosagem , Simulação por Computador , Quimioterapia Assistida por Computador/métodos , Eletrocardiografia/efeitos dos fármacos , Sistema de Condução Cardíaco/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Humanos , Agonistas do Receptor de Serotonina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA