Skip to main content
Erschienen in: Pediatric Cardiology 5/2023

Open Access 10.04.2023 | Review

Grafts and Patches: Optimized but Not Optimal Materials for Congenital Heart Surgery

verfasst von: Armin Darius Peivandi, Sven Martens, Boulos Asfour, Sabrina Martens

Erschienen in: Pediatric Cardiology | Ausgabe 5/2023

Abstract

A variety of materials are available for the surgery of children with congenital heart defects. In addition to growth-related mismatch, degeneration of the material in particular frequently leads to reoperation. Therefore, the choice of conduits and patches should be made carefully. This article provides an overview of the most commonly implanted materials in pediatric cardiac surgery.
Structural changes can be detected in all available materials. Depending on the age at implantation and the site of implantation, the extent and time course of material degeneration vary. Autologous material is still the gold standard in reconstructive surgery. Biological materials have largely replaced artificial materials in clinical use.
The search for the ideal material continues. In pediatric cardiac surgery, there are only optimized but no optimal materials.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Complex congenital heart defects usually require surgical treatment in infancy or early childhood. In many cases, either human (autologous, homologous), animal (bovine, porcine, equine), or artificial (DACRON®, DuPont Corporation, Wilmington, USA or GORE-TEX®, W.L. Gore & Associates, Inc., Newark, Delaware, USA) materials are used for reconstructive procedures in pediatric cardiac surgery. These materials exhibit varying durability depending on the implantation site and surgical technique. For example, patches (pericardial patches) degenerate differently than conduits (conduits with and without valves, such as Goretex). Degeneration of a variety of these materials results in frequent reoperations in childhood.
This review focuses on most commonly used materials that are mainly commercially available. The authors are fully aware of tissue-engineered center-specific modified patches, conduits, and valves [1]. Due to the heterogeneity of these materials, however, they cannot be covered in depth as part of this review.

Clinical Background

The prevalence of congenital heart defects is approximately 1% of live births. While ventricular and atrial septal defects are generally the most common congenital heart malformations (VSD (48.9%), ASD (17%)), the PAN study identified tetralogy of Fallot (TOF) (2.5%) and transposition of the great arteries (2.2%) as the most common complex cyanotic heart defects [2]. In most of these cardiac defects, correction is performed without planned reoperation. In some children, however, reoperations are required as part of multilevel therapy or because of growth-related mismatch, and in others, foreign material must be replaced, due to calcifications, pseudo-aneurysm formation, neo-atherosclerosis like lesions, and degeneration of collagen and elastic fibers [3, 4]. Children with congenital heart defects usually receive pediatric cardiological care and regular follow-up; as adults, they are referred to specialized outpatient clinics. In addition to regular physical examinations, echocardiography is a central component of imaging diagnostics. Degenerative changes can also be assessed by MRI and CT [5].
In the following, the problem of frequent reoperations in childhood mainly due to material degeneration is exemplified by the treatment options of TOF patients:
TOF describes the presence of the following four components: pulmonary stenosis, right ventricular hypertrophy, ventricular septal defect, and overriding aorta [6].
Today, surgical TOF correction is usually performed within the first year of life [6, 7]. The operation includes closure of the VSD and reconstruction of the right ventricular outflow tract (RVOT) [8]. Depending on the anatomic circumstances of the individual case (pulmonary stenosis, pulmonary atresia, absent pulmonary valve, size of the pulmonary valve annulus, coronary artery anatomy), surgical management of the RVOT may vary between infundibular myectomy, commissurotomy, patch expansion (transannular patch), and complete reconstruction with a conduit [69].
In most cases, valve-sparing surgery or transannular patchplasty are the preferred surgical options. However, reconstructive surgery with conduits is sometimes unavoidable.
Homografts (human prostheses from a donor bank) or Contegra® grafts (bovine jugular veins) are used for RVOT reconstruction. Degeneration of such valved prostheses due to calcification or fibrous deposits leads to stenosis and insufficiency, which may ultimately lead to early reoperation [3]. However, degenerative developments are not the only cause for graft replacement. In this context, somatic outgrowths leading to functional stenosis should also be mentioned.
No matter the cause, frequent reoperations in childhood result in an increased risk of surgery and a negative impact on the quality of life (physical, psychological, and overall). Withdrawal behavior, attention deficits, and externalization problems have been identified as psychological consequences [10].

Overview of Commonly Used Materials in Pediatric Cardiac Surgery

Table 1 provides an overview of some of the most commonly used materials in congenital heart surgery.
Table 1
Conduits, heart valves, and patches commonly used in pediatric cardiac surgery (random order, without claiming completeness)
Conduits
  Cardiac homografts
Tissue Banks (e.g., European Homograft Bank)
  Contegra® graft
Medtronic plc, Dublin, Ireland
  Hancock® conduit
Medtronic plc, Dublin, Ireland
  Freestyle™ prosthesis
Medtronic plc, Dublin, Ireland
  Biopulmonic conduit
Biointegral Surgical, Inc., Mississauga, ON
Heart valves
  Prosthetic heart valves
• Open Pivot by Medtronic
• SJM Regent™ by Abbott
• On-X by CryoLife
  Biological heart valves
• Perimount Magna Ease by Carpentier-Edwards
• SJM Trifecta™ by Abbott
Patches
  Autologous pericardium
  Bovine pericardial patches
• CardioCel patch by LeMaitre Vascular
• Peri-Guard® by LaMed
  Porcine pericardial patch
Curved NoReact Patch by BioIntegral
  Porcine submucosa patch
CorPatch® by CorMatrix® Cardiovascular
  Equine pericardial patch
Matrix Patch™ by Autotissue
  Artificial patches
GORE-TEX® by Gore Medical
DACRON® by DuPont Corporation

Conduits

Cardiac homografts are human grafts obtained from the left or right ventricular outflow tract of donors. Homografts are usually processed in specialized tissue banks where they are available on demand. While there are a number of officially accredited tissue banks in Europe (e.g., Hannover, Berlin, Bad Oeynhausen, Barcelona), many centers like ours rely on the European Homograft Bank in Brussels.
In 1989, the European Homograft Bank was founded as an internationally cooperating and non-profit organization [11]. Aortic and/or pulmonary explants are systematically processed there [12]. After standardized morphological assessment, professional preparation, and triple antibiotic treatment (vancomycin, lincomycin, polymyxin B), cryopreservation is performed [13].
The grafts are available to cardiac surgeons from all over Europe and can be ordered as needed (elective surgery or emergency surgery).
Due to the declining donor numbers and the resulting lack of available tissue in small sizes (10% of requests do not receive a positive vote) [14], alternative prostheses are increasingly gaining access to the medical device market. In this context, the Contegra® prosthesis occupies an exposed position.
The Contegra® graft (Medtronic plc, Dublin, Ireland), available since the 1990s, is a bovine jugular vein containing a tri-leaflet valve and processed in glutaraldehyde. In contrast to the homograft, the industrially produced Contegra® is often available in small sizes (diameter 12–22 mm, length 12–15 cm) [15].
In addition to the conduits described in detail above, further alternatives commercially available on the market must be mentioned. These include the Hancock® conduit (porcine aortic valve sewn into a woven conduit) [16, 17], the Freestyle prosthesis (porcine aortic root) (both (Medtronic plc, Dublin, Ireland)) [18, 19], or the BioPulmonalConduit (BioIntegral Surgical, Mississauga, Canada) [20].

Prosthetic and Biological Heart Valves

If possible, heart valve replacement is avoided in younger children. The first surgical attempt is usually rather reconstructive surgery of the valves. However, in cases where valve replacement is necessary (failed reconstruction attempts, teenagers), valve replacement is mainly performed using mechanical valves. Nowadays, these valves are double-wing heart valve prostheses. Many manufacturers have launched their products in the past. Common examples include but are not limited to Medtronic Open Pivot [21], St. Jude Medical Regent™ [22] and On-X. The last mentioned prosthesis was introduced especially for pediatric use as this valve requires lower Warfarin levels [23].
All mechanical valve prostheses carry bleeding risks as they require life-long anticoagulation. On the contrary, biological prostheses are almost exclusively used in pulmonary position due to their limited durability and to avoid thrombosis under low-pressure physiology. In the RVOT, e.g., Carpentier-Edwards Perimount Magna Ease [24, 25] or St. Jude Medical Trifecta™ prostheses [26, 27] can be used.

Patches

Pericardial patches currently used in pediatric cardiac surgery are described below:
Autologous pericardium is obtained from the same patient during surgery and implanted (if necessary, a brief treatment with 0.625% glutaraldehyde for 20 min and subsequent careful irrigation is performed) [28]. It currently represents the gold standard for reconstructive surgery, e.g., reconstruction of aortic valve leaflets [29].
The use of human material is limited, especially in re-do surgery when autologous pericardium cannot be used any more. In former days, fixation with glutaraldehyde was routinely performed, but studies now suggest a negative impact on tissue durability [30]. Non-glutaraldehyde-fixed pericardium is less pliable for surgical use. As a result, other biological materials have become popular. They mimic human pericardium but do not resemble the exact structure of their human counterparts.
The most common alternative to autologous pericardium is bovine pericardium. This material is produced by various manufacturers. The company LeMaitre Vascular (Sulzbach, Germany) offers the CardioCel patch, which has undergone a special anticalcification process (removal of cell particles and nucleic acids and minimization of glutaraldehyde content) [31]. According to the manufacturer, a special tear resistance characterizes other bovine patches from LaMed (Oberhaching, Germany). They are described as acellular [32].
Equine patches are also available (Matrix Patch™, Autotissue, Berlin, Germany). According to the manufacturer, they are made of decellularized horse pericardium (patches with low DNA content) [33].
Also of animal origin is the BioIntegral Curved NoReact Patch (BioIntegral Surgical, Mississauga, Canada), a curved porcine pericardial patch [34].
While pericardium is the most commonly used biomaterial for patch manufacturing, it is not the only one: the CorPatch® (CorMatrix® Cardiovascular, Roswell, Georgia, USA) is made from porcine small intestinal submucosa. In a porcine ischemic heart disease model, the material improved functional recovery of the myocardium [35]. In addition to the use of this material for epicardial infarct repair in adult patients, its performance has also been studied in children with congenital heart defects: No ingrowth of native cardiac tissue was detected after 21 months when the patch was used as a hemi-Fontan baffle [36].
Artificial materials have increasingly taken a back seat in pediatric reconstructions due to the multitude of biological alternatives. In earlier days, DACRON or GORE-TEX® were the materials that were mainly used. As thromboembolic complications along with short-term infections became evident in DACRON patches [37], this material was gradually replaced. Furthermore, tissue processing technology had improved and made biological materials more attractive to use. GORE-TEX®, a polytetrafluoroethylene patch that is also known in the textile industry for its durability, is used almost exclusively as a pericardial patch in chest closure for adhesion prophylaxis.

Degeneration as a Cause of Reoperations–Clinic and Histology

Degenerative changes are observed in all materials. They may occur at different time points after implantation and vary in their severity. When becoming apparent, they lead to reoperations. Macroscopic findings of explants from reoperations are shown in Fig. 1. Figure 2 shows inflammation and calcification as the most common cause of implant failure, in addition to growth-related mismatch, which inevitably leads to reoperations, too.
Implantation of homografts is still the strategy of choice in patients requiring reconstruction, e.g., of the RVOT [38]. Pulmonary homografts show very good mid-term results: the rate of reoperation in the first 8 years after implantation was only 7.8% in a recently published study (mean age of patients 26.1 ± 13.6 years) [39]. However, pulmonary homografts can present with clinical insufficiencies and obstruction, which may lead to graft replacement within 4–6 years in some patients [15]. Aortic homografts in pulmonary position are also prone to fibro-calcification, especially in patients younger than 3 years. This may be due to the lower elastin content of aortic homografts [40, 41].
Fiore et al. published their surgical results in infants and young children < 2 years of age and reported that 59% of their patients required reoperation due to high-grade stenosis and/or pulmonary insufficiencies [41]. Thus, the youngest patients are particularly affected by the problem of reoperation. Histologically, homograft failure is caused by intimal hyperplasia on the one hand [42], and calcifications, ossifications, and chronic immune reactions in the adventitia on the other hand [43, 44].
As a good surgical alternative to homografts, the Contegra® prosthesis has been generally accepted in the last 3 decades. This was mainly due to its easy surgical handling and availability in small sizes. A European multicenter study showed good 7-year results with respect to explant rates, endocarditis, stenosis, insufficiencies, and other events [45]. Compared to the homograft, the prosthesis shows a comparable hemodynamic performance. Its natural morphology has a positive influence on the surgical outcome [15, 46].
Nevertheless, there are also critical voices regarding the use of Contegra® grafts: Doubled reoperation rates compared to homografts are described [47], as well as infections of the prosthesis and problems with high RV-LV pressures and with small sizes [48, 49]. In the latter, stenotic fibrotic membranes and neointimal proliferation at the distal anastomosis are repeatedly observed [4, 50, 51]. The formal pathomechanism of Contegra® degeneration is based on elastic degeneration with accompanying stiffness of the prosthesis as well as on intimal hyperplasia with neointimal calcifications and heterotopic ossifications [3]. The polarization microscopic image of Contegra® reveals structural alterations caused by its production process (Fig. 3).
Patches represent a more heterogeneous graft group because they are derived from different species. Furthermore, their surgical use is more diverse.
Autologous pericardium can be harvested from any patient who has not undergone a large number of previous operations before. For many years, glutaraldehyde treatment of the harvested pericardium brought the advantages of better surgical handling due to higher mechanical stability, at the expense of an increased tendency to calcification [52]. Currently, the once established fixation method has therefore been abandoned in many clinics.
Severe calcification is the main cause of clinical degeneration of bovine pericardium used as a transannular patch in Fallot correction. Pseudo-aneurysmal changes and consequent pulmonary valve insufficiency are also observed.
Severe calcification is the main cause of clinical degeneration of bovine pericardium, which is often used as a transannular patch in TOF correction. Pseudo-aneurysmal changes and consequent pulmonary valve regurgitation are also observed. However, it is not just the degenerative change of the patch itself that necessitates reoperation, but the clinical increase in pulmonary insufficiency and consecutive right heart strain.
When used in reconstructive surgery of the aortic valve, echocardiographically immobile, thickened tissue portions can be seen after a longer period of time. This may lead to stenosis [53]. Histological correlates are calcifications and connective tissue deposits [54].
Currently, there are no reliable randomized clinical trials on equine pericardium. The manufacturer’s website refers to case reports [33]. The use of equine pericardium is justified by animal studies in which neither negative structural changes nor calcifications could be detected [55]. A recently published retrospective cohort study showed first good short-term clinical results [56], although no systematic distinctions were made between implantation site and congenital heart defect.
Porcine patches complete the animal product line and show promising clinical results [57]. A large-scale histological evaluation is also pending.

Conclusions and Future Challenges

Degeneration can be observed clinically in both conduits and patches used in pediatric cardiac surgery. It often results in unavoidable reoperations. However, not only the material as such, but also the implantation site and the surgical technique, as well as the age of the child seem to have an influence on long-term outcome.
To date, the ideal material for use in pediatric cardiac surgery has not been identified [58] as only optimized but no optimal materials are available.
Large-scale prospective randomized studies and systematic histopathological workups are desirable. New emerging techniques such as 3D printing, computational modeling, and tissue engineering may help providing individualized treatment options with optimal geometrical and flow properties in future [59].

Declarations

Conflict of interest

The authors declare that no conflicts of interest exist.

Ethical Approval

For the investigation of explants shown as figures of this review, a positive vote of the responsible ethics committee (Ethics Committee of the Medical Association of Westphalia-Lippe and the Westphalian Wilhelms University, 2015-231-f-S) was granted.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Pädiatrie

Kombi-Abonnement

Mit e.Med Pädiatrie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Pädiatrie, den Premium-Inhalten der pädiatrischen Fachzeitschriften, inklusive einer gedruckten Pädiatrie-Zeitschrift Ihrer Wahl.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Lü WD, Zhang M, Wu ZS, Hu TH (2009) Decellularized and photooxidatively crosslinked bovine jugular veins as potential tissue engineering scaffolds. Interact Cardiovasc Thorac Surg 8:301–305PubMedCrossRef Lü WD, Zhang M, Wu ZS, Hu TH (2009) Decellularized and photooxidatively crosslinked bovine jugular veins as potential tissue engineering scaffolds. Interact Cardiovasc Thorac Surg 8:301–305PubMedCrossRef
2.
Zurück zum Zitat Lindinger A, Schwedler G, Hense HW (2010) Prevalence of congenital heart defects in newborns in Germany: results of the first registration year of the PAN Study (July 2006 to June 2007). Klin Padiatr 222:321–326PubMedCrossRef Lindinger A, Schwedler G, Hense HW (2010) Prevalence of congenital heart defects in newborns in Germany: results of the first registration year of the PAN Study (July 2006 to June 2007). Klin Padiatr 222:321–326PubMedCrossRef
3.
Zurück zum Zitat Peivandi AD, Seiler M, Mueller KM, Martens S et al (2019) Elastica degeneration and intimal hyperplasia lead to Contegra® conduit failure. Eur J Cardiothorac Surg 56:1154–1161PubMedCrossRef Peivandi AD, Seiler M, Mueller KM, Martens S et al (2019) Elastica degeneration and intimal hyperplasia lead to Contegra® conduit failure. Eur J Cardiothorac Surg 56:1154–1161PubMedCrossRef
4.
Zurück zum Zitat Schoenhoff FS, Loup O, Gahl B et al (2011) The Contegra bovine jugular vein graft versus the Shelhigh pulmonic porcine graft for reconstruction of the right ventricular outflow tract: a comparative study. J Thorac Cardiovasc Surg 141:654–661PubMedCrossRef Schoenhoff FS, Loup O, Gahl B et al (2011) The Contegra bovine jugular vein graft versus the Shelhigh pulmonic porcine graft for reconstruction of the right ventricular outflow tract: a comparative study. J Thorac Cardiovasc Surg 141:654–661PubMedCrossRef
5.
Zurück zum Zitat Diller GP, Orwat S, Vahle J et al (2020) German competence network for congenital heart defects investigators. Prediction of prognosis in patients with tetralogy of fallot based on deep learning imaging analysis. Heart 106:1007–1014PubMedCrossRef Diller GP, Orwat S, Vahle J et al (2020) German competence network for congenital heart defects investigators. Prediction of prognosis in patients with tetralogy of fallot based on deep learning imaging analysis. Heart 106:1007–1014PubMedCrossRef
7.
Zurück zum Zitat Al Habib HF, Jacobs JP, Mavroudis C et al (2010) Contemporary patterns of management of tetralogy of fallot: data from the Society of Thoracic Surgeons Database. Ann Thorac Surg 90:813–819PubMedCrossRef Al Habib HF, Jacobs JP, Mavroudis C et al (2010) Contemporary patterns of management of tetralogy of fallot: data from the Society of Thoracic Surgeons Database. Ann Thorac Surg 90:813–819PubMedCrossRef
8.
Zurück zum Zitat Schmid C, Asfour B (eds) (2009). Steinkopff Verlag, Darmstadt Schmid C, Asfour B (eds) (2009). Steinkopff Verlag, Darmstadt
9.
Zurück zum Zitat Sommer RJ, Hijazi ZM, Rhodes JF (2008) Pathophysiology of congenital heart disease in the adult: Part III: complex congenital heart disease. Circulation 117:1340–1350PubMedCrossRef Sommer RJ, Hijazi ZM, Rhodes JF (2008) Pathophysiology of congenital heart disease in the adult: Part III: complex congenital heart disease. Circulation 117:1340–1350PubMedCrossRef
10.
Zurück zum Zitat Areias ME, Cl P, Vieira PF et al (2014) Living with CHD: quality of life (QOL) in early adult life. Cardiol Young 24(2):60–65PubMedCrossRef Areias ME, Cl P, Vieira PF et al (2014) Living with CHD: quality of life (QOL) in early adult life. Cardiol Young 24(2):60–65PubMedCrossRef
11.
Zurück zum Zitat Jashari R, Goffin Y, Vanderkelen A et al (2010) European homograft bank: twenty years of cardiovascular tissue banking and collaboration with transplant coordination in Europe. Transplant Proc 42:183–189PubMedCrossRef Jashari R, Goffin Y, Vanderkelen A et al (2010) European homograft bank: twenty years of cardiovascular tissue banking and collaboration with transplant coordination in Europe. Transplant Proc 42:183–189PubMedCrossRef
12.
Zurück zum Zitat Jashari R, Van Hoeck B, Ngakam R et al (2013) Banking of cryopreserved arterial allografts in Europe: 20 years of operation in the European Homograft Bank (EHB) in Brussels. Cell Tissue Bank 14:589–599PubMedCrossRef Jashari R, Van Hoeck B, Ngakam R et al (2013) Banking of cryopreserved arterial allografts in Europe: 20 years of operation in the European Homograft Bank (EHB) in Brussels. Cell Tissue Bank 14:589–599PubMedCrossRef
13.
Zurück zum Zitat Jashari R, Goffin Y, Van Hoeck B et al (2010) Belgian and European experience with the European Homograft Bank (EHB) cryopreserved allograft valves–assessment of a 20 year activity. Acta Chir Belg 110:280–290PubMedCrossRef Jashari R, Goffin Y, Van Hoeck B et al (2010) Belgian and European experience with the European Homograft Bank (EHB) cryopreserved allograft valves–assessment of a 20 year activity. Acta Chir Belg 110:280–290PubMedCrossRef
14.
Zurück zum Zitat Goffin YA, Van Hoeck B, Jashari R et al (2000) Banking of cryopreserved heart valves in Europe: assessment of a 10-year operation in the European Homograft Bank (EHB). J Heart Valve Dis 9:207–214PubMed Goffin YA, Van Hoeck B, Jashari R et al (2000) Banking of cryopreserved heart valves in Europe: assessment of a 10-year operation in the European Homograft Bank (EHB). J Heart Valve Dis 9:207–214PubMed
15.
Zurück zum Zitat Brown JW, Ruzmetov M, Rodefeld MD et al (2006) Valved bovine jugular vein conduits for right ventricular outflow tract reconstruction in children: an attractive alternative to pulmonary homograft. Ann Thorac Surg 82:909–916PubMedCrossRef Brown JW, Ruzmetov M, Rodefeld MD et al (2006) Valved bovine jugular vein conduits for right ventricular outflow tract reconstruction in children: an attractive alternative to pulmonary homograft. Ann Thorac Surg 82:909–916PubMedCrossRef
17.
Zurück zum Zitat Carney JP, Zhang LM, Larson JJ et al (2017) The Hancock® valved conduit for right ventricular outflow tract reconstruction in sheep for assessing new devices. J Heart Valve Dis 26:472–480PubMed Carney JP, Zhang LM, Larson JJ et al (2017) The Hancock® valved conduit for right ventricular outflow tract reconstruction in sheep for assessing new devices. J Heart Valve Dis 26:472–480PubMed
19.
Zurück zum Zitat Novick WM, Anic D, Solf AL et al (2004) Medtronic freestyle valve for right ventricular reconstruction in pediatric Ross operations. Ann Thorac Surg 77:1711–1716PubMedCrossRef Novick WM, Anic D, Solf AL et al (2004) Medtronic freestyle valve for right ventricular reconstruction in pediatric Ross operations. Ann Thorac Surg 77:1711–1716PubMedCrossRef
21.
Zurück zum Zitat Van Nooten GJ, Caes F, Francois K, Missault L, Van Belleghem Y (2009) Fifteen years’ single-center experience with the ATS bileaflet valve. J Heart Valve Dis 18:445–452 Van Nooten GJ, Caes F, Francois K, Missault L, Van Belleghem Y (2009) Fifteen years’ single-center experience with the ATS bileaflet valve. J Heart Valve Dis 18:445–452
22.
Zurück zum Zitat Jun BH, Saikrishnan N, Yoganathan AP (2014) Micro particle image velocimetry measurements of steady diastolic leakage flow in the hinge of a St. Jude Medical® regentTM mechanical heart valve. Biomed Eng 42:526–540 Jun BH, Saikrishnan N, Yoganathan AP (2014) Micro particle image velocimetry measurements of steady diastolic leakage flow in the hinge of a St. Jude Medical® regentTM mechanical heart valve. Biomed Eng 42:526–540
23.
Zurück zum Zitat Puskas JD, Gerdisch M, Nichols D, Fermin L, Rhenman B, Kapoor D, Copeland J, Quinn R, Hughes GC, Azar H, McGrath M, Wait M, Kong B, Martin T, Douville EC, Meyer S, Ye J, Jamison WRE, Landvater L, Hagberg R, Trotter T, Armitage J, Askew J, Accola K, Levy P, Duncan D, Yanagawa B, Ely J, Graeve A (2018) Anticoagulation and antiplatelet strategies after On-X mechanical aortic valve replacement. J Am Coll Cardiol 71:2717–2726PubMedCrossRef Puskas JD, Gerdisch M, Nichols D, Fermin L, Rhenman B, Kapoor D, Copeland J, Quinn R, Hughes GC, Azar H, McGrath M, Wait M, Kong B, Martin T, Douville EC, Meyer S, Ye J, Jamison WRE, Landvater L, Hagberg R, Trotter T, Armitage J, Askew J, Accola K, Levy P, Duncan D, Yanagawa B, Ely J, Graeve A (2018) Anticoagulation and antiplatelet strategies after On-X mechanical aortic valve replacement. J Am Coll Cardiol 71:2717–2726PubMedCrossRef
24.
Zurück zum Zitat Lee C, Park CS, Lee CH, Kwak JG, Kim SJ, Shim WS, Song JY, Choi EY, Lee SY (2011) Durability of bioprosthetic valves in the pulmonary position: long-term follow-up of 181 implants in patients with congenital heart disease. J Thorac Cardiovasc Surg 142:351–358PubMedCrossRef Lee C, Park CS, Lee CH, Kwak JG, Kim SJ, Shim WS, Song JY, Choi EY, Lee SY (2011) Durability of bioprosthetic valves in the pulmonary position: long-term follow-up of 181 implants in patients with congenital heart disease. J Thorac Cardiovasc Surg 142:351–358PubMedCrossRef
25.
Zurück zum Zitat Kwak JG, Lee JR, Kim WH, Kim YJ (2010) Mid-term results of the Hancock II valve and Carpentier-Edward perimount valve in the pulmonary portion in congenital heart disease. Heart Lung Circ 19:243–246PubMedCrossRef Kwak JG, Lee JR, Kim WH, Kim YJ (2010) Mid-term results of the Hancock II valve and Carpentier-Edward perimount valve in the pulmonary portion in congenital heart disease. Heart Lung Circ 19:243–246PubMedCrossRef
26.
Zurück zum Zitat Gulack BC, Benrashid E, Jaquiss RD, Lodge AJ (2017) Pulmonary valve replacement with a trifecta valve is associated with reduced transvalvular gradient. Ann Thorac Surg 103:655–662PubMedCrossRef Gulack BC, Benrashid E, Jaquiss RD, Lodge AJ (2017) Pulmonary valve replacement with a trifecta valve is associated with reduced transvalvular gradient. Ann Thorac Surg 103:655–662PubMedCrossRef
27.
Zurück zum Zitat Bavaria JE, Desai ND, Cheung A, Petracek MR, Groh MA, Borger MA, Schaff HV (2014) The St. Jude medical trifecta aortic pericardial valve: results from a global, multicenter, prospective clinical study. J Thorac Cardiovasc Surg 147:590–597PubMedCrossRef Bavaria JE, Desai ND, Cheung A, Petracek MR, Groh MA, Borger MA, Schaff HV (2014) The St. Jude medical trifecta aortic pericardial valve: results from a global, multicenter, prospective clinical study. J Thorac Cardiovasc Surg 147:590–597PubMedCrossRef
28.
Zurück zum Zitat Sánchez DM, Gaitán DM, León AF et al (2007) Fixation of vascular grafts with increased glutaraldehyde concentration enhances mechanical properties without increasing calcification. ASAIO J 53:257–262PubMedCrossRef Sánchez DM, Gaitán DM, León AF et al (2007) Fixation of vascular grafts with increased glutaraldehyde concentration enhances mechanical properties without increasing calcification. ASAIO J 53:257–262PubMedCrossRef
29.
Zurück zum Zitat Vergnat M, Asfour B, Arenz C et al (2017) Contemporary results of aortic valve repair for congenital disease: lessons for management and staged strategy. Eur J Cardiothorac Surg 52:581–587PubMedCrossRef Vergnat M, Asfour B, Arenz C et al (2017) Contemporary results of aortic valve repair for congenital disease: lessons for management and staged strategy. Eur J Cardiothorac Surg 52:581–587PubMedCrossRef
30.
Zurück zum Zitat Poppenborg F, Martens S, Martens S (2022) The influence of glutaraldehyde on the microscopic structure of human pericardium. Cardiovasc Pathol 61:107457PubMedCrossRef Poppenborg F, Martens S, Martens S (2022) The influence of glutaraldehyde on the microscopic structure of human pericardium. Cardiovasc Pathol 61:107457PubMedCrossRef
35.
Zurück zum Zitat Mewhort HE, Turnbull JD, Satriano A et al (2016) Epicardial infarct repair with bioinductive extracellular matrix promotes vasculogenesis and myocardial recovery. J Heart Lung Transplant 35:661–670PubMedCrossRef Mewhort HE, Turnbull JD, Satriano A et al (2016) Epicardial infarct repair with bioinductive extracellular matrix promotes vasculogenesis and myocardial recovery. J Heart Lung Transplant 35:661–670PubMedCrossRef
36.
Zurück zum Zitat Nelson JS, Heider A, Si MS, Ohye RG (2016) Evaluation of explanted CorMatrix Intracardiac patches in children with congenital heart disease. Ann Thorac Surg 102:1329–1335PubMedCrossRef Nelson JS, Heider A, Si MS, Ohye RG (2016) Evaluation of explanted CorMatrix Intracardiac patches in children with congenital heart disease. Ann Thorac Surg 102:1329–1335PubMedCrossRef
37.
Zurück zum Zitat Walhout RJ, Braam RL, Schepens MA, Mulder BJ, Plokker HW (2010) Aortic aneurysm formation following coarctation repair by Dacron patch aortoplasty. Neth Heart J 18:376–377PubMedPubMedCentralCrossRef Walhout RJ, Braam RL, Schepens MA, Mulder BJ, Plokker HW (2010) Aortic aneurysm formation following coarctation repair by Dacron patch aortoplasty. Neth Heart J 18:376–377PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Romeo JLR, Mokhles MM, van de Woestijne P et al (2019) Long-term clinical outcome and echocardiographic function of homografts in the right ventricular outflow tract. Eur J Cardiothorac Surg 55:518–526PubMedCrossRef Romeo JLR, Mokhles MM, van de Woestijne P et al (2019) Long-term clinical outcome and echocardiographic function of homografts in the right ventricular outflow tract. Eur J Cardiothorac Surg 55:518–526PubMedCrossRef
39.
Zurück zum Zitat Van Dang S, Pavy C, Guimbretière G et al (2021) Results of large pulmonary homograft implantation for right ventricular outflow tract reconstruction. J Card Surg 36:442–448CrossRef Van Dang S, Pavy C, Guimbretière G et al (2021) Results of large pulmonary homograft implantation for right ventricular outflow tract reconstruction. J Card Surg 36:442–448CrossRef
40.
Zurück zum Zitat Christenson JT, Sierra J, Colina Manzano NE et al (2010) Homografts and xenografts for right ventricular outflow tract reconstruction: long-term results. Ann Thorac Surg 90:1287–1294PubMedCrossRef Christenson JT, Sierra J, Colina Manzano NE et al (2010) Homografts and xenografts for right ventricular outflow tract reconstruction: long-term results. Ann Thorac Surg 90:1287–1294PubMedCrossRef
41.
Zurück zum Zitat Fiore AC, Ruzmetov M, Huynh D et al (2010) Comparison of bovine jugular vein with pulmonary homograft conduits in children less than 2 years of age. Eur J Cardiothorac Surg 38:318–325PubMedCrossRef Fiore AC, Ruzmetov M, Huynh D et al (2010) Comparison of bovine jugular vein with pulmonary homograft conduits in children less than 2 years of age. Eur J Cardiothorac Surg 38:318–325PubMedCrossRef
42.
Zurück zum Zitat Wells WJ, Arroyo H Jr, Bremner RM et al (2002) Homograft conduit failure in infants is not due to somatic outgrowth. JTCVS 124:88–96 Wells WJ, Arroyo H Jr, Bremner RM et al (2002) Homograft conduit failure in infants is not due to somatic outgrowth. JTCVS 124:88–96
43.
Zurück zum Zitat Butany J, Ahluwalia MS, Nair V et al (2004) Cryopreserved pulmonary homograft: postimplant changes. Cardiovasc Pathol 13:59–61PubMedCrossRef Butany J, Ahluwalia MS, Nair V et al (2004) Cryopreserved pulmonary homograft: postimplant changes. Cardiovasc Pathol 13:59–61PubMedCrossRef
44.
Zurück zum Zitat Carr-White GS, Kilner PJ, Hon JK et al (2001) Incidence, location, pathology, and significance of pulmonary homograft stenosis after the Ross operation. Circulation 104(Suppl 1):I16-20PubMedCrossRef Carr-White GS, Kilner PJ, Hon JK et al (2001) Incidence, location, pathology, and significance of pulmonary homograft stenosis after the Ross operation. Circulation 104(Suppl 1):I16-20PubMedCrossRef
45.
Zurück zum Zitat Breymann T, Blanz U, Wojtalik MA et al (2009) European Contegra Multicentre Study: 7-year results after 165 valved bovine jugular vein graft implantations. Thorac Cardiov Surg 57:257–259CrossRef Breymann T, Blanz U, Wojtalik MA et al (2009) European Contegra Multicentre Study: 7-year results after 165 valved bovine jugular vein graft implantations. Thorac Cardiov Surg 57:257–259CrossRef
46.
Zurück zum Zitat Corno AF, Qanadli SD, Sekarski N et al (2004) Bovine valved xenograft in pulmonary position: medium-term follow-up with excellent hemodynamics and freedom from calcification. Ann Thorac Surg 78:1382–1388PubMedCrossRef Corno AF, Qanadli SD, Sekarski N et al (2004) Bovine valved xenograft in pulmonary position: medium-term follow-up with excellent hemodynamics and freedom from calcification. Ann Thorac Surg 78:1382–1388PubMedCrossRef
47.
Zurück zum Zitat Urso S, Rega F, Meuris B et al (2011) The Contegra conduit in the right ventricular outflow tract is an independent risk factor for graft replacement. Eur J Cardiothorac Surg 40:603–609PubMed Urso S, Rega F, Meuris B et al (2011) The Contegra conduit in the right ventricular outflow tract is an independent risk factor for graft replacement. Eur J Cardiothorac Surg 40:603–609PubMed
48.
Zurück zum Zitat Albanesi F, Sekarski N, Lambrou D et al (2014) Incidence and risk factors for Contegra graft infection following right ventricular outflow tract reconstruction: long-term results. Eur J Cardiothorac Surg 45:1070–1074PubMedCrossRef Albanesi F, Sekarski N, Lambrou D et al (2014) Incidence and risk factors for Contegra graft infection following right ventricular outflow tract reconstruction: long-term results. Eur J Cardiothorac Surg 45:1070–1074PubMedCrossRef
49.
Zurück zum Zitat Shebani SO, McGuirk S, Baghai M et al (2006) Right ventricular outflow tract reconstruction using Contegra® valved conduit: natural history and conduit performance under pressure. Eur J Cardiothorac Surg 29:397–405PubMedCrossRef Shebani SO, McGuirk S, Baghai M et al (2006) Right ventricular outflow tract reconstruction using Contegra® valved conduit: natural history and conduit performance under pressure. Eur J Cardiothorac Surg 29:397–405PubMedCrossRef
50.
Zurück zum Zitat Kadner A, Dave H, Stallmach T et al (2004) Formation of a stenotic fibrotic membrane at the distal anastomosis of bovine jugular vein grafts (Contegra) after right ventricular outflow tract reconstruction. J Thorac Cardiovasc Surg 127:285–286PubMedCrossRef Kadner A, Dave H, Stallmach T et al (2004) Formation of a stenotic fibrotic membrane at the distal anastomosis of bovine jugular vein grafts (Contegra) after right ventricular outflow tract reconstruction. J Thorac Cardiovasc Surg 127:285–286PubMedCrossRef
51.
Zurück zum Zitat Meyns B, Van Garsse L, Boshoff D et al (2004) The Contegra conduit in the right ventricular outflow tract induces supravalvular stenosis. J Thorac Cardiovasc Surg 128:834–840PubMedCrossRef Meyns B, Van Garsse L, Boshoff D et al (2004) The Contegra conduit in the right ventricular outflow tract induces supravalvular stenosis. J Thorac Cardiovasc Surg 128:834–840PubMedCrossRef
52.
Zurück zum Zitat Sinha P, Sinha P, Zurakowski D et al (2012) Effects of glutaraldehyde concentration, pretreatment time, and type of tissue (porcine versus bovine) on postimplantation calcification. J Thorac Cardiovasc Surg 143:224–227PubMedCrossRef Sinha P, Sinha P, Zurakowski D et al (2012) Effects of glutaraldehyde concentration, pretreatment time, and type of tissue (porcine versus bovine) on postimplantation calcification. J Thorac Cardiovasc Surg 143:224–227PubMedCrossRef
53.
Zurück zum Zitat Nordmeyer S, Kretzschmar J, Murin P et al (2019) ADAPT-treated pericardium for aortic valve reconstruction in congenital heart disease: histological analysis of a series of human explants. Eur J Cardiothorac Surg 56:1170–1177PubMedCrossRef Nordmeyer S, Kretzschmar J, Murin P et al (2019) ADAPT-treated pericardium for aortic valve reconstruction in congenital heart disease: histological analysis of a series of human explants. Eur J Cardiothorac Surg 56:1170–1177PubMedCrossRef
54.
Zurück zum Zitat Nordmeyer S, Murin P, Schulz A et al (2018) Results of aortic valve repair using decellularized bovine pericardium in congenital surgery. Eur J Cardiothorac Surg 54:986–992PubMedCrossRef Nordmeyer S, Murin P, Schulz A et al (2018) Results of aortic valve repair using decellularized bovine pericardium in congenital surgery. Eur J Cardiothorac Surg 54:986–992PubMedCrossRef
55.
Zurück zum Zitat Dohmen PM, da Costa F, Lopes SV et al (2014) Successful implantation of a decellularized equine pericardial patch into the systemic circulation. Med Sci Monit Basic Res 20:1–8PubMedPubMedCentralCrossRef Dohmen PM, da Costa F, Lopes SV et al (2014) Successful implantation of a decellularized equine pericardial patch into the systemic circulation. Med Sci Monit Basic Res 20:1–8PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Elassal AA, Al-Radi OO, Zaher ZF et al (2021) Equine pericardium: a versatile alternative reconstructive material in congenital cardiac surgery. J Cardiothorac Surg 16:110PubMedPubMedCentralCrossRef Elassal AA, Al-Radi OO, Zaher ZF et al (2021) Equine pericardium: a versatile alternative reconstructive material in congenital cardiac surgery. J Cardiothorac Surg 16:110PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Yildrim Ö, Bakhshaliyev S, Kilercik H et al (2019) Early results of ring-reinforced conduit and curved porcine patch in Sano-Norwood procedure. J Card Surg 34:279–284CrossRef Yildrim Ö, Bakhshaliyev S, Kilercik H et al (2019) Early results of ring-reinforced conduit and curved porcine patch in Sano-Norwood procedure. J Card Surg 34:279–284CrossRef
58.
Zurück zum Zitat Cleuziou J (2018) Is there any progress in the search for the ideal conduit for reconstruction of the right ventricular outflow tract in young children? Transl Pediatr 7:226–228PubMedPubMedCentralCrossRef Cleuziou J (2018) Is there any progress in the search for the ideal conduit for reconstruction of the right ventricular outflow tract in young children? Transl Pediatr 7:226–228PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Manavitehrani I, Ebrahimi P, Yang I et al (2019) Current challenges and emergent technologies for manufacturing artificial right ventricle to pulmonary artery (RV-PA) cardiac conduits. Cardiovasc Eng Technol 10:205–215PubMedCrossRef Manavitehrani I, Ebrahimi P, Yang I et al (2019) Current challenges and emergent technologies for manufacturing artificial right ventricle to pulmonary artery (RV-PA) cardiac conduits. Cardiovasc Eng Technol 10:205–215PubMedCrossRef
Metadaten
Titel
Grafts and Patches: Optimized but Not Optimal Materials for Congenital Heart Surgery
verfasst von
Armin Darius Peivandi
Sven Martens
Boulos Asfour
Sabrina Martens
Publikationsdatum
10.04.2023
Verlag
Springer US
Erschienen in
Pediatric Cardiology / Ausgabe 5/2023
Print ISSN: 0172-0643
Elektronische ISSN: 1432-1971
DOI
https://doi.org/10.1007/s00246-023-03153-6

Weitere Artikel der Ausgabe 5/2023

Pediatric Cardiology 5/2023 Zur Ausgabe

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

TAVI versus Klappenchirurgie: Neue Vergleichsstudie sorgt für Erstaunen

21.05.2024 TAVI Nachrichten

Bei schwerer Aortenstenose und obstruktiver KHK empfehlen die Leitlinien derzeit eine chirurgische Kombi-Behandlung aus Klappenersatz plus Bypass-OP. Diese Empfehlung wird allerdings jetzt durch eine aktuelle Studie infrage gestellt – mit überraschender Deutlichkeit.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.