Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Artif Organs ; 44(10): 1061-1066, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32216106

ABSTRACT

The aim of the present study is to report our long-term experience with the HeartWare Ventricular Assist System (HVAD). Between July 2009 and February 2018, a total of 103 patients (mean age 50.0 ± 14.4, range 28-74 years; 22 females) received HVAD implantation in a single center institution. A total of 26 (25.4%) patients were in cardiogenic shock preoperatively and received extracorporeal life support (ECLS) prior to HVAD implantation. The aim of left ventricular assist device (LVAD) implantation was bridge to transplantation (BT) in 59 (57.3%), destination therapy (DT) in 28 (27.2%), and bridge to decision in 16 (15.5%). There were 211.1 total patient years of support. Mean survival was 2.05 ± 2.14 years. Kaplan-Meier analysis showed an overall survival rate of 69.7%, 56.7%, 46.0%, and 25.0% at 1, 2, 4, and 8 years, respectively. A total of 23 patients (22.3%) died during the hospital stay. Of them 65.2% (15 patients) were preoperatively in cardiogenic shock (INTERMACS 1). Sub-analysis of the BT patients showed a mean survival of 2.45 ± 2.29 years with a survival rate of 85.1%, 75.1%, 67.2%, and 44.8% at 1, 2, 4, and 8 years, respectively. Among them, 20 patients received heart transplantation on follow-up. Mean survival of DT patients was 2.18 ± 1.91 years with a survival rate of 67.9%, 49.0%, and 25.1% at 1, 2, and 4 years, respectively. At latest follow-up in September 2018, 26 patients (25.24%) were still on LVAD. A total of five patients completed 6 years on LVAD, of them two were supported over 8 years. The most common adverse event reported was gastrointestinal bleeding requiring rehospitalization (0.161 EPPY). A total of 19 patients reported disabling stroke. Pump thrombosis was diagnosed in six patients (5.8%) (0.02 EPPY), of them four patients underwent pump exchange. To the best of our knowledge, this is the longest experience with HVAD reported so far. Patients supported with an HVAD show a satisfactory long-term survival. Further multicenter evaluations are needed to confirm these single-center results.


Subject(s)
Gastrointestinal Hemorrhage/epidemiology , Heart Failure/surgery , Heart Transplantation/statistics & numerical data , Heart-Assist Devices/statistics & numerical data , Postoperative Hemorrhage/epidemiology , Adult , Aged , Female , Follow-Up Studies , Gastrointestinal Hemorrhage/etiology , Heart Failure/mortality , Heart-Assist Devices/adverse effects , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Postoperative Hemorrhage/etiology , Retrospective Studies , Survival Rate , Treatment Outcome
2.
Thorac Cardiovasc Surg ; 66(4): 295-300, 2018 06.
Article in English | MEDLINE | ID: mdl-28201842

ABSTRACT

BACKGROUND: Minimally invasive surgical access through limited sternotomy reduces trauma and morbidity in cardiosurgical patients. However, until now, it is not the standard access for aortic root replacement. This study details our clinic's experience with minimally invasive implantation of valved conduits through partial upper sternotomy and the comparison to conventional full median sternotomy. METHODS: Between January 2012 and March 2016, a total of 187 patients underwent aortic root replacement with valved conduits in our department. Minimally invasive access through partial upper sternotomy (group A) was performed in 33 patients (9 female, 24 male; mean age: 55.68 ± 13.24 years). Four of these patients received concomitant mitral and tricuspid valve interventions. The results were compared with similar procedures through conventional approach (group B): 25 patients (7 female, 18 male; mean age: 59.09 ± 12.32 years). RESULTS: In all 33 cases of minimally invasive access and 25 cases of conventional approach, aortic root replacement was successful. Operative times were as follows (in minutes; groups A and B)-cardiopulmonary bypass: 166.12 ± 40.61 and 162.88 ± 45.89; cross-clamp time: 122.24 ± 27.42 and 113.44 ± 22.57, respectively. In both groups, two patients needed postoperative reexploration due to secondary bleeding. One multimorbid patient suffered from postoperative stroke and died on the ninth postoperative day due to heart failure. The observed operation times and clinical results after minimally invasive surgery are comparable to conduit implantation through full median sternotomy. CONCLUSIONS: Partial upper sternotomy is a feasible access for safe aortic root replacement with valved conduits. Nevertheless, minimally invasive aortic root replacement is a challenging operative procedure.


Subject(s)
Aortic Aneurysm/surgery , Aortic Valve Insufficiency/surgery , Aortic Valve/abnormalities , Blood Vessel Prosthesis Implantation/methods , Heart Valve Diseases/surgery , Heart Valve Prosthesis Implantation/methods , Sternotomy/methods , Adult , Aged , Aortic Aneurysm/diagnostic imaging , Aortic Aneurysm/mortality , Aortic Aneurysm/physiopathology , Aortic Valve/diagnostic imaging , Aortic Valve/physiopathology , Aortic Valve/surgery , Aortic Valve Insufficiency/diagnostic imaging , Aortic Valve Insufficiency/mortality , Aortic Valve Insufficiency/physiopathology , Bicuspid Aortic Valve Disease , Blood Vessel Prosthesis Implantation/adverse effects , Cardiopulmonary Bypass , Feasibility Studies , Female , Heart Failure/etiology , Heart Valve Diseases/diagnostic imaging , Heart Valve Diseases/mortality , Heart Valve Diseases/physiopathology , Heart Valve Prosthesis Implantation/adverse effects , Hospital Mortality , Humans , Male , Middle Aged , Minimally Invasive Surgical Procedures , Operative Time , Postoperative Hemorrhage/surgery , Risk Factors , Sternotomy/adverse effects , Stroke/etiology , Time Factors , Treatment Outcome
3.
Artif Organs ; 41(5): 418-423, 2017 May.
Article in English | MEDLINE | ID: mdl-27862075

ABSTRACT

Left ventricular assist device (LVAD) implantation is a well-established therapy to support patients with end-stage heart failure. However, the operative procedure is associated with severe trauma. Third generation LVADs like the HeartWare assist device (HeartWare, Inc., Framingham, MA, USA) are characterized by enhanced technology despite smaller size. These devices offer new minimally invasive surgical options. Tricuspid regurgitation requiring valve repair is frequent in patients with the need for mechanical circulatory support as it is strongly associated with ischemic and nonischemic cardiomyopathy. We report on HeartWare LVAD implantation and simultaneous tricuspid valve reconstruction through minimally invasive access by partial upper sternotomy to the fifth left intercostal space. Four male patients (mean age 51.72 ± 11.95 years) suffering from chronic heart failure due to dilative (three patients) and ischemic (one patient) cardiomyopathy and also exhibiting concomitant tricuspid valve insufficiency due to annular dilation underwent VAD implantation and tricuspid valve annuloplasty. Extracorporeal circulation was established via the ascending aorta, superior vena cava, and right atrium. In all four cases the LVAD implantation and tricuspid valve repair via partial median sternotomy was successful. During the operative procedure, no conversion to full sternotomy was necessary. One patient needed postoperative re-exploration because of pericardial effusion. No postoperative focal neurologic injury was observed. New generation VADs are advantageous because of the possibility of minimally invasive implantation procedure which can therefore minimize surgical trauma. Concomitant tricuspid valve reconstruction can also be performed simultaneously through partial upper sternotomy. Nevertheless, minimally invasive LVAD implantation is a challenging operative technique.


Subject(s)
Heart Failure/surgery , Heart Ventricles/surgery , Heart-Assist Devices , Minimally Invasive Surgical Procedures/methods , Prosthesis Implantation/methods , Tricuspid Valve Insufficiency/surgery , Tricuspid Valve/surgery , Adult , Heart Failure/complications , Heart Failure/physiopathology , Heart Ventricles/physiopathology , Humans , Male , Middle Aged , Sternotomy/methods , Tricuspid Valve/physiopathology , Tricuspid Valve Insufficiency/complications , Tricuspid Valve Insufficiency/physiopathology
4.
Thorac Cardiovasc Surg ; 64(6): 483-6, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26720706

ABSTRACT

A minority of patients on left ventricular assist devices (LVADs) display myocardial recovery raising the question of device explantation. We report a series of seven LVAD patients with nonischemic cardiomyopathy allowing explantation of the VAD. In case of four patients we overview follow-up periods of 4 to 10 years. Remarkably, in three cases device explantation was considered beyond 1 year of LVAD support. Explantation was finally performed after 20 months and more on the device. In light of the scarcity of donor organs we conclude that patients on LVAD support should be evaluated for possible weaning from the device on a regular basis.


Subject(s)
Cardiomyopathies/therapy , Device Removal , Heart Failure/therapy , Heart-Assist Devices , Pregnancy Complications, Cardiovascular/therapy , Prosthesis Implantation/instrumentation , Ventricular Function, Left , Adolescent , Adult , Cardiomyopathies/diagnosis , Cardiomyopathies/physiopathology , Female , Heart Failure/diagnosis , Heart Failure/physiopathology , Heart Transplantation , Humans , Magnetic Resonance Imaging , Male , Pregnancy , Pregnancy Complications, Cardiovascular/diagnosis , Pregnancy Complications, Cardiovascular/physiopathology , Prosthesis Design , Prosthesis Implantation/adverse effects , Recovery of Function , Time Factors , Tissue Donors/supply & distribution , Treatment Outcome , Waiting Lists , Young Adult
5.
J Cardiothorac Vasc Anesth ; 30(3): 627-31, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26460277

ABSTRACT

OBJECTIVES: Right heart failure still occurs in up to 20% of patients after implantation of a left ventricular assist device (LVAD). One treatment option for these patients is the implantation of a temporary right ventricular assist device (RVAD). Experimental data suggest that non-pulsatile perfusion of the lungs is associated with an increased rate of pulmonary hemorrhage. The aim of this study was to determine the incidence of pulmonary bleeding complications in these patients. DESIGN: Observational study. SETTING: Single center, university hospital. PARTICIPANTS: This study included patients undergoing LVAD implantation for end-stage heart failure and subsequent implantation of a temporary right ventricular support system. INTERVENTIONS: In this study, 25 patients who underwent LVAD and additional temporary RVAD implantation were screened for pulmonary bleeding complications. MEASUREMENTS AND MAIN RESULTS: The mean Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS) level at the time of LVAD implantation was 2.84. All patients experienced severe right ventricular failure (tricuspid annular plane systolic excursion [TAPSE], 10.16±26.3 mm) and severe pulmonary hypertension (right atrial [RA] pressure, 56.21±15.58 mmHg). Average duration of right ventricular support was 11.12±7.20 days, with right ventricular support being administered to 14 patients for more than 7 days. Seventeen patients were weaned successfully from right ventricular support after a mean support duration of 5 days. Five patients developed pulmonary bleeding complications, diagnosed using computed tomography scan and bronchoscopy. All bleeding occurred after postoperative day 7 and was associated with RVAD flow of more than 4 L/min within 24 hours before bleeding occurred. CONCLUSIONS: The data presented in this study suggested that right ventricular support for more than 7 days and a blood flow greater than 4 L/min were associated with pulmonary bleeding complications. This should be taken into consideration when temporary right ventricular support after LVAD implantation is planned.


Subject(s)
Heart Failure/surgery , Heart-Assist Devices/adverse effects , Hemorrhage/etiology , Lung Diseases/etiology , Adult , Aged , Bronchoscopy , Extracorporeal Membrane Oxygenation/adverse effects , Extracorporeal Membrane Oxygenation/methods , Female , Hemorrhage/diagnosis , Humans , Hypertension, Pulmonary/complications , Lung Diseases/diagnosis , Male , Middle Aged , Postoperative Period , Prosthesis Implantation/adverse effects , Prosthesis Implantation/methods , Tomography, X-Ray Computed
6.
J Cardiothorac Vasc Anesth ; 30(3): 619-26, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27321789

ABSTRACT

OBJECTIVES: Depending on the pre-existing condition of the right ventricle (RV), left ventricular assist device (LVAD) implantation may have a detrimental effect on RV function, subsequently leading to right heart failure. This study details the authors' experience with perioperative mechanical RV support in patients with biventricular impairment but primarily scheduled for isolated LVAD implantation. DESIGN: Retrospective study. SETTING: Two center study, university hospital. PARTICIPANTS: This study included LVAD recipients with preoperative biventricular impairment who received an additional right ventricular assist device (RVAD) after a failed weaning attempt from cardiopulmonary bypass due to acute RV failure. INTERVENTIONS: Outcomes of 25 patients who underwent LVAD and unplanned temporary RVAD implantation were analyzed. MEASUREMENTS AND MAIN RESULTS: All patients experienced significant preoperative RV impairment (tricuspid annular plane systolic excursion: 10.2±26.3 mm; right atrium pressure: 17.9±10.4 mmHg) and pulmonary hypertension (pulmonary artery pressure: 54.8±25.7 mmHg). In 15 patients, additional tricuspid valve annuloplasty was performed. Mean duration of temporary RVAD support was 11.1±7.2 days. In 23 patients (92%), the RVAD was removed successfully. None of the patients developed RV failure after RVAD removal. Hospital survival and the 1-year survival rate of the study group were 68% and 56%, respectively. CONCLUSIONS: The results of perioperative RVAD support in LVAD recipients with biventricular dysfunction are encouraging. Temporary RVAD support allows an already compromised RV to become attuned to the hemodynamic conditions after LVAD implantation. This strategy provides patients with preoperative impaired RV function a high likelihood to permanently undergo LVAD support only.


Subject(s)
Heart Failure/prevention & control , Heart-Assist Devices , Prosthesis Implantation/methods , Adult , Aged , Cardiopulmonary Bypass , Extracorporeal Membrane Oxygenation/methods , Female , Heart Failure/etiology , Humans , Male , Middle Aged , Perioperative Care/methods , Prosthesis Implantation/adverse effects , Retrospective Studies , Treatment Outcome , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Right/physiopathology
7.
J Artif Organs ; 19(3): 305-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27003430

ABSTRACT

Peripartum cardiomyopathy (PPCM) challenges different disciplines including gynecologists, cardiologists and cardiac surgeons. It is a severe cardiac failure occurring at the end of pregnancy or within the first months after delivery. Urgent intervention is necessary to stabilize hemodynamics with appropriate medication and mechanical assist support. Data of 4 patients with PPCM at a mean age of 28.2 years were analyzed retrospectively. Echocardiography was used to evaluate cardiac function. Despite varying courses of PPCM all women received left ventricular assist devices (LVAD) since their hemodynamics could not be stabilized by medical treatment. Mean gestational week at delivery was 31.5 ± 4.9. Left ventricular ejection fractions of all patients were severely impaired (17.8 ± 3.5 %) before LVAD implantations. After long-term mechanical support (282 and 417 days, respectively) 2 patients were successfully transplanted. Two other females could be weaned from LVAD therapy after 944 and 612 days, respectively. LVAD explanations were performed according to a standardized weaning protocol. Myocardial recovery was confirmed by regular echocardiography, spiroergometry and right heart catheterization. Mechanical assist device support is a surgical strategy either as bridge to transplantation or as bridge to recovery for patients with PPCM.


Subject(s)
Cardiomyopathy, Dilated/therapy , Heart Transplantation , Heart-Assist Devices , Peripartum Period , Adult , Cardiomyopathy, Dilated/physiopathology , Female , Hemodynamics/physiology , Humans , Pregnancy , Retrospective Studies , Treatment Outcome
8.
Artif Organs ; 39(12): 998-1004, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26011007

ABSTRACT

Poor survival has been demonstrated after ventricular assist device (VAD) implantation for Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS) profile 1 and 2 patients compared with more stable levels. However, risk factors within this high-risk cohort have not been determined so far. The aim of the present study was to identify risk factors associated with this very high mortality rate. Between February 1993 and January 2013, 298 patients underwent VAD implantation in our institution. One hundred nine patients were in INTERMACS level 1 and 49 patients were in INTERMACS level 2 and were therefore defined as hemodynamically critical (overall 158 patients). Assist devices implanted were: HVAD HeartWare n = 18; Incor n = 11; VentrAssist n = 2; DeBakey n = 22; and pulsatile systems n = 105. After cumulative support duration of 815.35 months, Kaplan-Meier analysis revealed a survival of 63.9, 48.8, and 40.3% at 1, 6, and 12 months, respectively. Cox regression analyses identified age > 50 (P = 0.001, odds ratio [OR] 2.48), white blood cell count > 13.000/µL (P = 0.01, OR 2.06), preoperative renal replacement therapy (P = 0.001, OR 2.63), and postcardiotomy failure (P < 0.001, OR 2.79) as independent predictors of mortality. Of note, last generation VADs were not associated with significantly better 6-month survival (P = 0.59). Patients without the aforementioned risk factors could yield a survival of 79.2% at 6 months. This single-center experience shows that VAD implantation in hemodynamically unstable patients generally results in poor early outcome, even in third-generation pumps. However, avoiding the aforementioned risk factors could result in improved outcome.


Subject(s)
Heart-Assist Devices , Hemodynamics , Shock, Cardiogenic/therapy , Ventricular Function, Left , Adult , Aged , Contraindications , Critical Illness , Female , Germany , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Patient Selection , Proportional Hazards Models , Risk Assessment , Risk Factors , Shock, Cardiogenic/diagnosis , Shock, Cardiogenic/etiology , Shock, Cardiogenic/mortality , Shock, Cardiogenic/physiopathology , Treatment Outcome , Young Adult
9.
J Artif Organs ; 18(4): 291-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26044781

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a rare but life-threatening side effect of heparin therapy. It is a demanding therapeutic challenge in patients undergoing left ventricular assist device (LVAD) implantation. We present our experience with LVAD implantation under extracorporeal life support (ECLS) in patients suffering from HIT. Seven patients (mean age 54.0 ± 16.7 years, 1 female, 6 male patients) suffering from acute heart failure were stabilized with ECLS. Under heparin therapy, they all showed a sudden decrease of mean platelet count (maximum 212.6 ± 41.5 tsd/µl; minimum 30.7 ± 13.1 tsd/µl). Due to the clinical suspicion of HIT anticoagulation was switched from heparin to argatroban (aPTT 70-80 s.). No improvement of cardiac function could be detected under ECLS, so LVAD implantation was indicated. We performed LVAD implantation under ECLS. During LVAD implantation under argatroban (aPTT of 50-60 s.) one patient developed massive intraventricular thrombus formations, so the device had to be removed. In 6 cases, we could successfully perform LVAD implantation under argatroban with a higher aPTT of 70-80 s and modified operative strategy. Four patients needed postoperative re-exploration because of bleeding complications. Perioperative management of LVAD patients under argatroban anticoagulation is very difficult. We were able to minimize the perioperative risk for thrombosis with a target aPTT of 70-80 s. To keep the phase of stasis within the device as short as possible we anastomosed the VAD outflow graft to the ascending aorta first before connecting the device to the apex. However, postoperative bleeding complications are frequent.


Subject(s)
Anticoagulants/adverse effects , Extracorporeal Circulation , Heart Failure/therapy , Heart-Assist Devices , Heparin/adverse effects , Prosthesis Implantation , Thrombocytopenia/chemically induced , Acute Disease , Adult , Aged , Arginine/analogs & derivatives , Cohort Studies , Female , Heart Failure/complications , Humans , Male , Middle Aged , Pipecolic Acids/therapeutic use , Sulfonamides , Thrombocytopenia/surgery , Thrombosis/etiology
10.
ESC Heart Fail ; 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769653

ABSTRACT

AIMS: Technological advances and the current shortage of donor organs have contributed to an increase in the number of left ventricular assist device (LVAD) implantations in patients with end-stage heart failure. Demographic change and medical progress might raise the number of these patients, resulting in a further increase in the number of LVAD implantations. The aim of this study was to evaluate the long-term costs of LVAD therapy and identify diagnoses resulting in expensive stays. METHODS AND RESULTS: In this retrospective analysis of prospectively collected data, all patients after implantation of a second- or third-generation LVAD by 31 March 2022 were included. In addition to demographic and survival data, revenues and case mix points were determined for each patient. Of the 163 patients included, 75.5% were male. The mean age at LVAD implantation was 52 ± 14 years. The mean survival was 1458 ± 127 days. During follow-up, the total inpatient treatment time per patient was 70 ± 87 days. The average duration of outpatient treatment was 55.1%, based on the total duration of support. The average revenue per patient for the implant stay was $193 192.35 ± $111 801.29, for inpatient readmissions $52 068.96 ± $116 630.00, and for outpatient care $53 195.94 ± $62 363.53. CONCLUSIONS: LVAD implantation in patients with end-stage heart failure leads to improved survival but a significant increase in treatment costs. Further multi-centre studies are necessary in order to be able to assess the effects of long-term LVAD treatment on the healthcare system.

11.
Eur J Cardiothorac Surg ; 62(1)2022 06 15.
Article in English | MEDLINE | ID: mdl-35024851

ABSTRACT

OBJECTIVES: Gastrointestinal bleeding in patients with continuous-flow left ventricular assist devices (CF-LVAD) impairs quality of life and increases hospitalization rate. The Utah Bleeding Risk Score (UBRS) has been created to predict gastrointestinal bleeding (GIB) in patients on left ventricular assist device. We here aimed to externally validate UBRS on our cohort of CF-LVAD patients. METHODS: Utah Bleeding Risk Score was calculated, GIB events summarized on follow-up and patients stratified into 3 risk groups. Predictive ability of UBRS was examined at 3 years and during whole support time and person time incidence rates correlated to UBRS. In a sub-analysis, single effects of UBRS variables on freedom from GIB were assessed. RESULTS: A total of 111 CF-LVAD patients were included. The median UBRS was 2 (3-1). Receiver operating characteristic curve analysis showed an area under the curve of 0.519 (P = 0.758, 95% confidence interval = 0.422-0.615) at 3 years and an area under the curve of 0.515 (P = 0.797, 95% confidence interval = 0.418-0.611) during whole support time. No significant difference was observed in UBRS between bleeders and non-bleeders (P = 0.80). No significant difference in freedom from GIB was observed (P3 years = 0.7; Psupport-time = 0.816) and no independent significance regarding the freedom from bleeding at 3 years for any variable was observed. Coronary artery disease was associated with higher risk of GIB beyond the 3rd year (P = 0.048). CONCLUSIONS: UBRS was not able to predict GIB and therefore not applicable in our cohort of patients. Coronary artery disease could lead to a higher risk for GIB during support time. An additional validation in a larger cohort is advisable.


Subject(s)
Coronary Artery Disease , Heart Failure , Heart-Assist Devices , Coronary Artery Disease/complications , Gastrointestinal Hemorrhage/epidemiology , Gastrointestinal Hemorrhage/etiology , Heart-Assist Devices/adverse effects , Humans , Quality of Life , Retrospective Studies , Risk Factors , Utah/epidemiology
13.
J Artif Organs ; 12(3): 160-5, 2009.
Article in English | MEDLINE | ID: mdl-19894089

ABSTRACT

Implantation of left ventricular assist device (LVAD) as a bridge to recovery or transplantation is a widely accepted treatment modality. Preexisting organ dysfunction is thought to unfavorably affect patient survival after implantation of a ventricular assist device (VAD). We present our experience using extracorporeal membrane oxygenation (ECMO) in patients with cardiogenic shock to stabilized organ function prior to LVAD implantation. Between September 2006 and March 2008, five patients in cardiogenic shock with preexisting organ dysfunction (impaired liver and kidney function) were supported with ECMO before LVAD implantation. ECMO-LVAD interval was 8 +/- 4 days. All patients were transferred to a LVAD. At the LVAD implantation time, glutamic-oxaloacetic transaminase (GOT) decreased from 206.25 +/- 106.93 Ul (-1) to 70.6 +/- 32.9 U l(-1), glutamic-pyruvic transaminase (GPT) decreased from 333.5 +/- 207.3 U l(-1) to 77.8 +/- 39.7 U l(-1), and creatinine decreased from 2.2 +/- 0.9 mg dl(-1) to 1.2 +/- 0.2 mg dl(-1). One patient died while on LVAD support due to not device related sepsis. One patient received successful heart transplantation. Overall survival was 80%. In all patients, we removed the ECMO 3 days after LVAD implantation. After removal of the ECMO there was no right heart failure. ECMO support can immediately stabilize circulation and provide organ perfusion in patients with cardiogenic shock. After improvement of organ function, LVAD implantation can be performed successfully in this patient collective. To avoid right ventricular failure, the ECMO should not be removed at the time of LVAD implantation, and used as a right ventricular support for the immediate postoperative period.


Subject(s)
Extracorporeal Membrane Oxygenation , Heart-Assist Devices , Shock, Cardiogenic/therapy , Ventricular Dysfunction, Right/therapy , Adult , Female , Humans , Male , Middle Aged , Perioperative Care , Prosthesis Implantation , Ventricular Dysfunction, Left/therapy
14.
Eur Heart J Cardiovasc Imaging ; 19(5): 532-543, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28977417

ABSTRACT

Aim: Presence and consequent extent of infection in patients on continuous-flow left ventricular assist devices (CF-LVADs) can be challenging with the current diagnostic tools. The present study sought to demonstrate the diagnostic power of 18F-Fluorodeoxyglucose-Positron-Emission Tomography/Computed Tomography (18F-FDG PET/CT) in detecting infection in patients supported with CF-LVAD. Background: The present study sought to demonstrate the diagnostic power of 18F-fluorodeoxyglucose-positron-emission tomography/computed tomography (18F-FDG PET/CT) in detecting infection in patients supported with CF-LVAD. Methods and results: Between July 2009 and April 2016, 61 PET/CT examinations were performed in 47 patients (median age 64.13 years, IQR 18.77) supported with a CF-LVAD. PET/CT assessments were performed qualitatively and quantitatively at three different levels: at the piercing site of driveline (first level), along the intracorporeal course of driveline (second level), and around the device (third level). Final diagnosis of LVAD infection was prospectively performed and was based upon microbiological samples taken at hospital admission, during the surgical revision/transplantation and recurrence of symptoms on long-term follow-up. At last follow-up a total of 40 (65.57%) final diagnoses of LVAD-infection could be ascertained. Matching the final diagnosis with the PET/CT assessments the sensitivity, specificity, and positive and negative predictive value were 90.0, 71.4, 85.71, and 78.94%, respectively. Level sub-analyses of SUV max showed an optimal discriminator power for levels 1 and 2 (AUC of level 1-0.824, P < 0.001; AUC of level 2-0.849, P < 0.001, respectively). At the third level semi-quantitative analysis showed poor discriminator power (AUC 0.589, P = 0.33). Qualitative visual analysis instead indicated a trend toward significance (P = 0.07). Conclusions: Quantitative 18F-FDG PET/CT is an optimal diagnostic tool in detecting superficial and deep driveline infections. However, diagnostic accuracy with regard to the diagnosis of pump housing infection is limited. Here, clinical and qualitative PET/CT analyses must be better considered.


Subject(s)
Fluorodeoxyglucose F18 , Heart-Assist Devices/adverse effects , Positron Emission Tomography Computed Tomography/methods , Prosthesis-Related Infections/diagnostic imaging , Prosthesis-Related Infections/epidemiology , Radiographic Image Enhancement , Academic Medical Centers , Adult , Aged , Anti-Bacterial Agents/therapeutic use , Cohort Studies , Combined Modality Therapy , Female , Heart Transplantation/methods , Humans , Male , Middle Aged , Prevalence , Prognosis , Prosthesis-Related Infections/microbiology , Prosthesis-Related Infections/therapy , ROC Curve , Retrospective Studies , Risk Assessment , Severity of Illness Index , Survival Rate , Treatment Outcome
15.
J Heart Valve Dis ; 16(2): 126-31, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17484458

ABSTRACT

BACKGROUND AND AIM OF THE STUDY: The study aim was to evaluate the efficacy of left atrial (LA) size reduction combined with radiofrequency (RF) ablation in the treatment of continuous atrial fibrillation (AF), by comparative analysis of the outcomes of patients undergoing RF ablation with and without LA size reduction. METHODS: A total of 46 patients with continuous AF and cardiac disease underwent cardiac surgery and RF ablation alone (group I, n = 20) or combined with LA size reduction (group II, n = 26). Patients were followed for three years postoperatively, with evaluation of cardiac rhythm, neurological complications, LA size (by echocardiography) and atrial contractility. RESULTS: At three years after surgery, sinus rhythm (SR) was restored in 61.1% and 70% of patients in groups I and II, respectively. Mean LA diameter was reduced from 60 +/- 15 mm to 57 +/- 5 mm in group I, and from 69 +/- 19 mm to 55 +/- 6 mm in group II. The overall three-year survival was 90% in group I, and 88.5% in group II. Three-year freedom from stroke was 88.9% and 86.2% in groups I and II, respectively. Two patients in each group received transvenous permanent pacemaker implantation. Atrial contractility was recovered in all patients with stable SR. CONCLUSION: LA size reduction improves SR conversion rate after RF ablation for continuous AF in patients undergoing concomitant cardiac surgery.


Subject(s)
Atrial Fibrillation/surgery , Cardiac Surgical Procedures , Catheter Ablation , Endocardium/surgery , Adult , Aged , Atrial Fibrillation/mortality , Atrial Fibrillation/physiopathology , Atrial Function, Left , Combined Modality Therapy , Female , Follow-Up Studies , Heart Atria/physiopathology , Heart Atria/surgery , Humans , Male , Middle Aged , Postoperative Period , Prospective Studies , Survival Rate , Time Factors , Treatment Outcome
16.
Ann Thorac Surg ; 101(1): 87-94; discussion 94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26433521

ABSTRACT

BACKGROUND: The current study sought to demonstrate the advantages offered by fluorine 18-fluorodeoxyglucose ((18)F-FDG) positron emission tomography/computed tomography (PET/CT) in patients supported with continuous-flow left ventricular assist devices (CF-LVADs) in detecting infection and the consequent effect on clinical decisions. METHODS: Between April 2009 and September 2013, 40 PET examinations were performed in 31 patients (78.1% men; mean age, 51.0 ± 14.9 years) supported with a CF-LVAD. In group A (19 examinations in 14 patients), PET/CT was performed to detect infectious focus in patients without external signs of driveline involvement but with at least two of the following infection signs: recurrent fever, positive blood culture, or elevated infectious indicators. In group B (21 examinations in 17 patients), PET/CT aimed to assess the internal extension of infection in patients with external signs of driveline infection. RESULTS: In 50% of the cases of the patients in group A, abnormal (18)F-FDG uptake (9 patients) was related to VAD components. Matching the results with the final diagnosis, we reported 9 true-positive, 8 true-negative, no false-negative, and 2 false-positive findings. New information unrelated to VAD was found in 9 patients (50%): pneumonia in 3, colon diverticulitis in 3, sternal dehiscence in 1, paravertebral abscess in 1, and erysipelas in 1. In group B, superficial abnormal (18)F-FDG uptake was found at the piercing site of the driveline in 2 patients, deeper extension of infection along the driveline in 10, initial involvement of the pump housing in 2, and full involvement of the device in 4. These findings contributed to changing the clinical management in 84.2% of group A patients and in 85.7% of group B patients: 16 patients were scheduled for urgent transplantation, 2 underwent surgical revision of the driveline, 7 required prolonged antibiotic therapy, and 3 underwent colonoscopy. CONCLUSIONS: This single-center experience highlights the diagnostic value of PET/CT in detecting the localization and internal extension of infection to internal VAD components. Moreover, this information notably influences the therapeutic management.


Subject(s)
Fluorodeoxyglucose F18 , Heart-Assist Devices , Positron-Emission Tomography/methods , Prosthesis-Related Infections/diagnosis , Tomography, X-Ray Computed/methods , Anti-Bacterial Agents/therapeutic use , Device Removal , Female , Follow-Up Studies , Heart Failure/therapy , Humans , Male , Middle Aged , Prosthesis-Related Infections/therapy , Radiopharmaceuticals , Reproducibility of Results , Retrospective Studies
17.
Exp Clin Transplant ; 13(5): 485-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25232871

ABSTRACT

Right (systemic) ventricular failure is well described after a Mustard procedure for transposition of the great arteries. Sixty-one percent of patients have been reported to show moderate-to-severe dysfunction of the systemic ventricle, 25 years after repair of transposition of the great arteries by a Mustard procedure. In such cases, heart transplant often seems to be the only option. When severe heart failure with hemodynamic instability develops, the only way to bridge these patients to a transplant is implantation of a mechanical circulatory support system. Because of the special anatomic features of the right ventricle, the implantation procedure can be challenging. Here, we report the case of a patient who underwent a left ventricular assist device implant owing to severe heart failure, 36 years after undergoing a Mustard procedure.


Subject(s)
Arterial Switch Operation/adverse effects , Heart Failure/therapy , Heart Transplantation , Heart-Assist Devices , Transposition of Great Vessels/surgery , Ventricular Dysfunction, Left/therapy , Ventricular Function, Left , Adult , Female , Heart Failure/diagnosis , Heart Failure/etiology , Heart Failure/physiopathology , Humans , Prosthesis Design , Severity of Illness Index , Time Factors , Transposition of Great Vessels/diagnosis , Treatment Outcome , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology , Waiting Lists
18.
Ann Thorac Surg ; 73(1): 233-8; discussion 238-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11834015

ABSTRACT

BACKGROUND: Activation of the complement system and polymorphonuclear neutrophilic leukocytes plays a major role in mediating reperfusion injury after lung transplantation. We hypothesized that early interference with complement activation would reduce lung reperfusion injury after transplantation. METHODS: Unilateral left lung autotransplantation was performed in 6 sheep. After hilar stripping the left lung was flushed with Euro-Collins solution and preserved for 2 hours in situ at 15 degrees C. After reperfusion the right main bronchus and pulmonary artery were occluded, leaving the animal dependent on the reperfused lung (reperfused group). C1-esterase inhibitor group animals (n = 6) received 200 U/kg body weight of C1-esterase inhibitor as a short infusion, half 10 minutes before, the other half 10 minutes after reperfusion. Controls (n = 6) underwent hilar preparation only. Pulmonary function was assessed by alveolar-arterial oxygen difference and pulmonary vascular resistance. The release of beta-N-acetylglucosaminidase served as indicator of polymorphonuclear neutrophilic leukocyte activation. Extravascular lung water was an indicator for pulmonary edema formation. Biopsy specimens were taken from all groups 3 hours after reperfusion for light and electron microscopy. RESULTS: In the reperfused group, alveolar-arterial oxygen difference and pulmonary vascular resistance were significantly elevated after reperfusion. All animals developed frank alveolar edema. The biochemical marker beta-N-acetylglucosaminidase showed significant leukocyte activation. In the C1-esterase inhibitor group, alveolar-arterial oxygen difference, pulmonary vascular resistance, and the level of polymorphonuclear neutrophilic leukocyte activation were significantly lower. CONCLUSIONS: Treatment with C1-esterase inhibitor reduces reperfusion injury and improves pulmonary function in this experimental model.


Subject(s)
Complement C1 Inactivator Proteins/therapeutic use , Lung Transplantation , Reperfusion Injury/prevention & control , Animals , Complement Activation/physiology , Female , Hypertonic Solutions , Leukocytes, Mononuclear/physiology , Lung/physiology , Lung Transplantation/adverse effects , Lung Transplantation/physiology , Models, Animal , Organ Preservation Solutions , Oxygen/metabolism , Sheep , Vascular Resistance
19.
J Heart Valve Dis ; 12(4): 469-74, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12918849

ABSTRACT

BACKGROUND AND AIM OF THE STUDY: Left atrial enlargement is a risk factor for the development of atrial fibrillation (AF). Large atrial size increases thromboembolic risk and reduces the success rate of cardioversion. The study aim was to evaluate if left atrial size reduction affects cardiac rhythm in patients with chronic AF undergoing mitral valve surgery. METHODS: Twenty-seven patients were analyzed prospectively. The left atrial incision was extended to the left inferior pulmonary vein. Left atrial size reduction was achieved by closure of the left atrial appendage from inside with a double running suture. The same suture plicated the left lateral atrial wall to the roof of the left pulmonary vein inflow and the inferior atrial wall. The atrial septum was plicated by placing stitches of the closing suture line across the fossa ovalis. Rhythm, neurological complications, cardioversion, anticoagulation and anti-arrhythmic medication were evaluated at one year postoperatively and at recent follow up (mean 40 +/- 15 months). RESULTS: At discharge, five patients (19%) were in sinus rhythm (SR). At one year postoperatively, SR was restored in 17 patients (63%), but five (19%) reported episodes of arrhythmia and AF persisted in 10 (37%). At recent follow up, four patients had died and three were lost to follow up. Among 20 patients examined, 13 (65%) had SR but six reported episodes of arrhythmia and AF persisted in seven (35%). LA diameter was significantly reduced, from 60.2 +/- 9.8 mm preoperatively to 44.5 +/- 7.0 mm at one year after surgery. CONCLUSION: The addition of left atrial size reduction to mitral valve surgery is technically simple, and was effective in 63% of patients with chronic AF, restoring predominant SR. In order to influence pathogenetic factors other than size, additional ablative steps may further increase the SR conversion rate. Size reduction may also improve the outcome of other ablative approaches.


Subject(s)
Cardiopulmonary Bypass , Heart Atria/pathology , Heart Atria/surgery , Mitral Valve/surgery , Adult , Aged , Aged, 80 and over , Amiodarone/therapeutic use , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation , Cardiac Pacing, Artificial , Chronic Disease , Cohort Studies , Digitoxin/therapeutic use , Echocardiography , Electric Countershock , Female , Follow-Up Studies , Heart Conduction System/drug effects , Heart Conduction System/pathology , Heart Valve Diseases/therapy , Humans , Male , Middle Aged , Mitral Valve/pathology , Postoperative Complications/etiology , Postoperative Complications/mortality , Postoperative Complications/therapy , Sotalol/therapeutic use , Survival Analysis , Treatment Outcome , Verapamil/therapeutic use
20.
Interact Cardiovasc Thorac Surg ; 18(6): 732-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24632426

ABSTRACT

OBJECTIVES: Patients undergoing cardiac surgery presenting with chronic obstructive pulmonary disease (COPD) have a higher 30-day mortality risk. In these patients, pulmonary dysfunction linked to an inflammatory response is frequent after cardiac operations using cardiopulmonary bypass (CPB), which causes pulmonary hypoperfusion. We hypothesize that selective pulmonary perfusion (sPP) of the lungs leads to a reduction of pulmonary inflammation and a better clinical outcome. METHODS: Fifty-nine COPD patients (forced expiratory volume in 1 s/vital capacity <70%) undergoing cardiac surgery procedures (coronary artery bypass grafting 64%, valve 14%) were block-randomized to sPP (venous blood, temperature 2°C, 4 l) or standard CPB (28/28). The primary end-point of the study was to evaluate the effect of pulmonary perfusion on gas exchange by measuring alveolar-arterial oxygen gradient. The surrogate end-points were inflammatory response, intensive care unit (ICU) stay, time on respirator (TOR) and major adverse cardiac and cerebrovascular events. A cytokine assay for interleukin-1ß, IL-6, IL-10, tumour necrosis factor-α (TNF-α) and polymorphonuclear elastase was performed with peripheral blood at different time-points [(t1) pre-CPB, (t2) end of CPB, (t3) 3 h, (t4) 24 h, (t5) 48 h postoperatively]. Repeated-measure analysis of variance and non-parametric statistics were used to assess the between-group and during time differences. RESULTS: The two groups proved comparable for perioperative variables. Serum cytokines were not different in the two groups throughout the study (P > 0.05 at single time-points), but as a function of time, the markers of the inflammatory response increased after CBP (P < 0.05 pre-CPB to 24 h). Clinical end-points were statistically comparable in both groups, but with a trend towards a shorter TOR (72 ± 159 h/106 ± 193 h) and ICU stay (3.9 ± 7.2 days/5.5 ± 9.2 days) in the sPP group despite a slightly longer time on extracorporeal circulation (120 vs 158 min). CONCLUSIONS: These results indicate a non-significant trend that repeated hypothermic lung perfusion with venous blood during CPB may have a protective effect on the lungs. A multicentre study design and larger cohort seem necessary to demonstrate the benefits of sPP more clearly.


Subject(s)
Coronary Artery Bypass , Coronary Artery Disease/surgery , Heart Valve Diseases/surgery , Lung/blood supply , Perfusion/methods , Pneumonia/prevention & control , Pulmonary Disease, Chronic Obstructive/complications , Aged , Biomarkers/blood , Cardiopulmonary Bypass/adverse effects , Coronary Artery Bypass/adverse effects , Coronary Artery Disease/complications , Coronary Artery Disease/diagnosis , Female , Forced Expiratory Volume , Germany , Heart Valve Diseases/complications , Heart Valve Diseases/diagnosis , Humans , Hypothermia, Induced , Inflammation Mediators/blood , Lung/physiopathology , Male , Middle Aged , Perfusion/adverse effects , Pneumonia/blood , Pneumonia/diagnosis , Pneumonia/etiology , Pneumonia/physiopathology , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Gas Exchange , Risk Factors , Time Factors , Treatment Outcome , Vital Capacity
SELECTION OF CITATIONS
SEARCH DETAIL