Retrospective Study Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Cardiol. Mar 26, 2022; 14(3): 177-186
Published online Mar 26, 2022. doi: 10.4330/wjc.v14.i3.177
Risk profiles and outcomes of patients receiving antibacterial cardiovascular implantable electronic device envelopes: A retrospective analysis
David A Woodard, Kent R Nilsson, Department of Cardiology, Piedmont Heart Institute, Athens, GA 30606, United States
Grace Kim, Kent R Nilsson, Department of Medicine, Augusta University-University of Georgia Medical Partnership, Athens, GA 30606, United States
ORCID number: David A Woodard (0000-0001-7046-911X); Grace Kim (0000-0002-8769-8864); Kent R Nilsson (0000-0001-8000-9686).
Author contributions: Woodard D led the conception, design, data collection and analysis, and drafting of the manuscript; Nilsson K and Kim G contributed their expertise to the analysis/interpretation of data and editing of the manuscript; and all authors accept accountability for the accuracy of this work, and drafted, revised, and approved the final version of the manuscript to be published within this journal.
Supported by Aziyo Biologics, Inc.
Institutional review board statement: This study was reviewed and approved by the Ethics Committee of the WIRB-Copernicus Group®.
Informed consent statement: A waiver of informed consent and HIPAA due to the retrospective nature of this study was obtained. This study was conducted in accordance with the ethical principles in the Declaration of Helsinki and conducted according to United States and international standards of Good Clinical Practice in accordance with applicable Federal regulations, International Council for Harmonization guidelines, and institutional research policies and procedures.
Conflict-of-interest statement: Woodard D is a consultant for Aziyo Biologics, Inc. The other authors have no relevant financial relationships to disclose.
Data sharing statement: The dataset supporting the conclusions of this article is available upon reasonable request to the corresponding author.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: David A Woodard, MD, Doctor, Department of Cardiology, Piedmont Heart Institute, Piedmont Athens Regional Medical Center 242 King Ave Suite 210, Athens, GA 30606, United States. davidwoodard92464@gmail.com
Received: December 17, 2021
Peer-review started: December 17, 2021
First decision: January 25, 2022
Revised: February 8, 2022
Accepted: March 3, 2022
Article in press: March 3, 2022
Published online: March 26, 2022

Abstract
BACKGROUND

Cardiovascular implantable electronic devices (CIEDs) are implanted in an increasing number of patients each year, which has led to an increase in the risk of CIED infection. Antibacterial CIED envelopes locally deliver antibiotics to the implant site over a short-term period and have been shown to reduce the risk of implant site infection. These envelopes are derived from either biologic or non-biologic materials. There is a paucity of data examining patient risk profiles and outcomes from using these envelope materials in the clinical setting and comparing these results to patients receiving no envelope with their CIED implantation.

AIM

To evaluate risk profiles and outcomes of patients who underwent CIED procedures with an antibacterial envelope or no envelope.

METHODS

After obtaining Internal Review Board approval, the records of consecutive patients who underwent a CIED implantation procedure by a single physician between March 2017 and December 2019 were retrospectively collected from our hospital. A total of 248 patients within this period were identified and reviewed through 12 mo of follow up. The CIED procedures used either no envelope (n = 57), a biologic envelope (CanGaroo®, Aziyo Biologics) that was pre-hydrated by the physician with vancomycin and gentamicin (n = 89), or a non-biologic envelope (Tyrx™, Medtronic) that was coated with a resorbable polymer containing the drug substances rifampin and minocycline by the manufacturer (n = 102). Patient selection for receiving either no envelope or an envelope (and which envelope to use) was determined by the treating physician. Statistical analyses were performed between the 3 groups (CanGaroo, Tyrx, and no envelope), and also between the No Envelope and Any Envelope groups by an independent, experienced biostatistician.

RESULTS

On average, patients who received any envelope (biologic or non-biologic) were younger (70.7 ± 14.0 vs 74.9 ± 10.6, P = 0.017), had a greater number of infection risk factors (81.2% vs 49.1%, P < 0.001), received more high-powered devices (37.2% vs 5.8%, P = 0.004), and were undergoing more reoperative procedures (47.1% vs 0.0%, P < 0.001) than patients who received no envelope. Between the two envelopes, biologic envelopes tended to be used more often in higher risk patients (84.3% vs 78.4%) and reoperative procedures (62.9% vs 33.3%) than non-biologic envelopes. The rate of CIED implant site pocket infection was low (any envelope 0.5% vs no envelope 0.0%) and was statistically equivalent between the two envelope groups. Other reported adverse events (lead dislodgement, lead or pocket revision, device migration or erosion, twiddler’s syndrome, and erythema/fever) were low and statistically equivalent between groups (biologic 2.2%, non-biologic 3.9%, no envelope 1.8%).

CONCLUSION

CIED infection rates for biologic and non-biologic antibacterial envelopes are similar. Antibacterial envelopes may benefit patients who are higher risk for infection, however additional studies are warranted to confirm this.

Key Words: Cardiovascular implantable electronic device envelope, Defibrillator, Extracellular matrix, Implantable cardioverter-defibrillator, Infection, Pacemaker

Core Tip: This retrospective study was performed to determine risk profiles and clinical outcomes of patients who underwent cardiovascular implantable electronic device (CIED) procedures with a biologic or non-biologic antibacterial envelope, or no envelope. A total of 248 patient records were reviewed containing 89 biologic, 102 non-biologic, and 57 no envelope patients. Pre-procedurally, patients who received any envelope (biologic or non-biologic) were at higher infection risk than patients who received no envelope. Biologic envelopes tended to be used more often in higher risk patients than non-biologic envelopes. The rate of CIED pocket infection was low and equivalent between the two envelopes.



INTRODUCTION

Expanding indications for cardiovascular implantable electronic devices (CIEDs) have increased the number of these devices that are implanted[1], but considering the common comorbidities seen in this patient population, complications such as infection are also increasing[2-4]. Reported infection rates of de novo CIED implantation range between 0.7%-4.6%, and can be as high as 7% for re-operations[5]. Thus, a better understanding of patient risk factors and available prophylactic techniques could potentially lower the risk of infection in this population[5-8]. CIED envelopes are intended to securely hold pacemakers or defibrillators when implanted in the body, and antibacterial CIED envelopes additionally provide short-term local antibiotic delivery which can reduce the risk of infection at the device implant site[9]. Available antibacterial CIED envelopes are either fabricated from biologic material (extracellular matrix hydrated with antibiotics by physician choice) or from non-biologic material (synthetic mesh coated with antibiotics by the manufacturer). The biologic envelope (CanGaroo®, Aziyo Biologics, Inc., Roswell, GA, United States) is made of decellularized extracellular matrix derived from porcine intestinal submucosa (SIS-ECM) which is rehydrated in solution for 1–2 min prior to use, whereas the non-biologic envelope (TYRX™, Medtronic PLC, Mounds View, MN, United States) is made from an absorbable synthetic substrate mesh coated with a bioresorbable polymer containing the drug substances rifampin and minocycline. Both envelopes have been reported to release antibiotics over a period of seven days in separate studies[10-13].

Although both envelopes have similar indications and antibiotic elution abilities, the material each envelope is created from may affect the biologic response upon implantation into the patient. Synthetic (non-biologic) absorbable and non-absorbable materials have been reported to initiate a strong foreign body reaction, resulting in chronic inflammation leading to hypovascular fibrotic tissue surrounding the implanted material[14-18], which a previously-marketed non-absorbable synthetic envelope leveraged to stabilize the electronic device upon implantation[19]. Conversely, ECM (the material that the biologic envelope is made from) has been shown to promote constructive remodeling and healthy tissue restoration[20-23]. Both biologic and non-biologic envelopes have been reported to support clinical infection prevention strategies[12,24-26].

This is an analysis of a retrospective, real-world study which assessed the risk profiles and clinical outcomes of patients who underwent a CIED procedure and received an antibacterial envelope (biologic or non-biologic), or no envelope (CARE Plus, NCT04351269). To our best knowledge, this study contains the first reporting of biologic and non-biologic antibacterial envelopes reported together in the clinical setting.

MATERIALS AND METHODS

Records of consecutive patients undergoing CIED procedures from a single center performed by a single physician between March 2017 and December 2019 were retrospectively reviewed for up to 12 mo of follow-up. The study protocol was reviewed and approved by an independent internal review board (IRB) [WIRB-Copernicus Group® (WCG)] prior to the chart review. A waiver of informed consent and HIPAA was obtained due to the retrospective nature of the study.

The study aimed to determine risk profiles and clinical outcomes of patients who were undergoing a CIED procedure and received either no envelope, a biologic envelope (CanGaroo®) hydrated by the implanting physician for 1 – 2 minutes with a vancomycin and gentamicin solution before implantation, or a non-biologic envelope (TYRX™) coated by the manufacturer with a bioresorbable polymer containing the drug substances rifampin and minocycline. The implanting physician made all decisions regarding device type, which envelope and envelope size was used, and biologic envelope hydration solution (if one was used). Aside from the pre-hydration of the biologic envelope, the implanting technique of both the biologic and non-biologic envelope was similar. The no envelope group’s CIED implantation procedure was identical to the envelope CIED implantation procedure, just without the use of an envelope. The pre- and post-operative protocol was the same for all 3 groups.

Information was extracted in detail from medical records, including medical history, infection risk factors, surgical details, and adverse events from the initial procedural visit out to 12 mo post-op. Infection risk factors were defined by previous literature[4,27,28] which identified elements that were significantly associated with increased risk for CIED infection, including renal insufficiency, diabetes, obesity, peripheral vascular disease, chronic obstructive pulmonary disease, congestive heart failure, malignancy, coronary artery disease, hypertension, chronic steroid use, oral systemic anticoagulants, malnutrition, smoking, the presence of two or more leads, pocket re-entry within 2 wk of the initial implant, prior device infection, and reoperative procedure. The number of risk factors was counted for each patient to examine the relative levels of infection risk between patient groups. Infection risk was categorized for each patient as lower risk (0–1 infection risk factors) or higher risk (2 or more risk factors), based on the quantity of established clinical risk factors present in each patient from above. An independent, biomedical statistician performed analyses between the 3 groups (CanGaroo, Tyrx, and no envelope), and also between the no envelope and any envelope groups by using means with standard deviations for continuous variables and counts with percentages for categorical variables. Continuous variables were checked for normality. Fisher’s exact tests were used when ≥ 1 expected cell counts were < 5, and Pearson chi-square tests were used for categorical variable comparisons when cell counts were ≥ 5. Statistical significance was set to a P < 0.05. SPSS version 26 (IBM, Armonk, NY, United States) was used for statistical analyses.

RESULTS

Among 248 enrolled patients who underwent CIED procedures, 191 (77%) received an envelope. These included 89 (46.6%) biologic and 102 (53.4%) non-biologic envelopes (Table 1).

Table 1 Comparison across cohorts.

Total
Biologic envelope
Non-biologic envelope
No envelope
1P value
Any envelope
2P value
(N = 248)(n = 89)(n = 102)( n = 57)( n = 191)
Age (yr, mean ± SD)71.6 ± 13.373.6 ± 13.368.2 ± 14.074.9 ± 10.60.00270.7 ± 14.00.017
BMI, mean ± SD29.9 ± 7.128.0 ± 6.231.0 ± 7.931.0 ± 6.30.00829.6 ± 7.30.206
BMI category0.0160.080
Underweight (< 18.5)5 (2.0%)2 (2.2%)2 (2.0%)1 (1.8%)4 (2.1%)
Normal (18.5 - < 25.0)54 (21.8%)30 (33.7%)19 (18.6%)5 (8.8%)49 (25.7%)
Overweight (25.0 - < 30.0)73 (29.4%)20 (22.5%)31 (30.4%)22 (38.6%)51 (26.7%)
Obese (30.0 - < 40.0)97 (39.1%)34 (38.2%)38 (37.3%)25 (43.9%)72 (37.7%)
Morbidly obese (40.0 ±)19 (7.7%)3 (3.4%)12 (11.8%)4 (7.0%)15 (7.9%)
Medical history
Heart failure106 (42.7%)41 (46.1%)49 (48.0%)16 (28.1%)0.03790 (47.1%)0.011
Systemic anticoagulant use99 (39.9%)43 (48.3%)40 (39.2%)16 (28.1%)0.05083 (43.5%)0.037
CIED device type0.0040.001
Pacemaker152 (61.3%)52 (58.4%)52 (51.0%)48 (84.2%)104 (54.5%)
CRT-P12 (4.8%)8 (9.0%)4 (3.9%)0 (0.0%)12 (6.3%)
ICD54 (21.8%)17 (9.1%)30 (29.4%)7 (12.3%)47 (24.6%)
S-ICD2 (0.8%)0 (0.0%)52 (51.0%)1 (1.8%)1 (0.5%)
CRT-D24 (9.7%)10 (11.2%)13 (12.7%)1 (1.8%)23 (12.0%)
N/A4 (1.6%) 2 (2.2%)2 (2.0%)0 (0.0%)0 (0.0%)
CIED category0.0060.004
Low-powered 164 (66.1%)60 (67.4%)56 (54.9%)48 (84.2%)116 (60.7%)
High-powered 80 (32.3%)27 (30.3%)44 (43.1%)9 (5.8%)71 (37.2%)
N/A4 (1.6%)2 (2.2%)2 (2.0%)0 (0.0%)4 (2.1%)
Procedure type< 0.001< 0.001
De novo158 (63.7%)33 (20.9% de novo)68 (43.0% de novo)57 (36.1% de novo)101 (63.9% de novo)
Re-operative90 (36.3%)56 (62.2% re-op)34 (37.8% re-op)0 (0.0% re-op)90 (100% re-op)
Infection risk factors< 0.001< 0.001
0-165 (26.2%)14 (15.7%)22 (21.6%)29 (50.9%)36 (18.8%)
≥ 2183 (73.8%)75 (84.3%)80 (78.4%)28 (49.1%)155 (81.2%)
Hematoma (total)6 (2.4%)5 (5.6%)1 (1.0%)0 (0.0%)0.0466 (3.0%)0.176
Requiring intervention6 (2.4%)5 (5.6%)1 (1.0%)0 (0.0%)6 (3.0%)
Infection
Pocket infection1 (0.4%)1 (1.1%)0 (0.0%)0 (0.0%)0.4081 (0.5%)0.584
Minor infection1 (0.4%)1 (1.1%)0 (0.0%)0 (0.0%)0.4081 (0.5%)0.584
Surgical procedure details

Patients who received high-powered devices, including implantable cardioverter defibrillators (ICDs) and cardiac resynchronization therapy (CRT) devices, were more likely to receive an envelope (P = 0.001) (Table 1). Patients undergoing reoperative procedures (generator changes, upgrades, other reoperative procedures such as lead or pocket revisions) received an envelope significantly more often than no envelope (100.0% vs 0.0%, P < 0.001) and tended to be more likely to receive a biologic than a non-biologic envelope (n = 56, 62.9% vs n = 34, 33.3%). Those with de novo implants tended to be more likely to receive a non-biologic envelope (n = 68, 66.6%) than a biologic envelope (n = 33, 37.1%).

Clinical characteristics and infection risk factors

Patients who received any envelope were younger on average (70.7 ± 14.0 vs 74.9 ± 10.6 years, P = 0.017) and had higher rates of comorbid risk factors such as heart failure (47.1% vs 28.1%, P = 0.011) and systemic anticoagulation (43.5% vs 28.1%, P = 0.037) than those who did not receive an envelope (Table 1). Patients with biologic envelopes tended to be somewhat older (mean 73.6 ± 13.3 vs 68.2 ± 14.0 years) and less overweight (22.5% vs 30.4%) than those with non-biologic envelopes. Differences in systemic anticoagulation among the 3 groups were statistically significant (biologic 48.3%, non-biologic 39.2%, no envelope 28.1%, P = 0.050). Patients who received any envelope had a significantly higher number of infection risk factors (≥ 2) than those with no envelope (81.2% vs 49.1%, P < 0.001), and biologic envelopes tended to be used more frequently for these higher risk patients (84.3% vs 78.4%).

Infection outcomes

Pocket infection rates were low (envelope 0.5%, no envelope 0.0%), with no significant difference between biologic and non-biologic envelopes (Table 1). Among the patients who received an envelope, one (0.5%) developed a major CIED infection (pocket infection), and one (0.5%) developed a minor CIED infection (superficial surgical site infection). However, the incidence of major or minor infection did not significantly differ between the 3 cohorts.

Other adverse events

Pocket hematoma (requiring surgical intervention) developed in 6 patients (2.4%): 5 patients (5.6%) with biologic envelopes, 1 patient (1.0%) with a non-biologic envelope, and 0 patients without an envelope (0.0%) (P = 0.046) (Table 1). However, there was no significant difference in hematoma between any envelope (3.0%) and no envelope (0.0%). There were no reported hematoma that led to infections in this study. Other adverse events included 3 Lead dislodgements (1 in the biologic group, 2 in the non-biologic group), 1 Lead revision (non-biologic group), 1 hemothorax (non-biologic group), and 1 site drainage (biologic group) in the envelope cohorts and erythema/fever in 1 patient in the no envelope cohort. Rates of adverse events other than pocket hematoma did not significantly differ among the 3 cohorts.

DISCUSSION

This retrospective study examined clinical profiles and outcomes of patients receiving CIEDs implanted with antibacterial biological envelopes hydrated with gentamicin and vancomycin (biologic envelopes), CIEDs implanted with synthetic (non-biologic) antibacterial envelopes, and CIEDs with no envelope. Non-biologic antibacterial envelopes have been previously shown in a large, randomized study to reduce infection risk in patients who are at increased risk for CIED infection[12]. To the best of our knowledge, this is the first reporting of clinical outcomes from using either biologic or non-biologic antibacterial envelopes, or no envelope within the same dataset.

Patient selection for envelope use

Patient selection by the implanting physician is reflected in the study findings. Envelopes were selected significantly more often for younger patients, patients undergoing device replacement procedures, high-powered device implantations, those on systemic anticoagulation, patients with heart failure, and patients with 2 or more risk factors for CIED infection. Treatment preferences can be observed by envelope usage for at-risk patients who may benefit most from the local delivery of antibiotics to their CIED implant site. Interestingly, there was no statistical difference in observed infection rates between the envelope and no envelope groups, even though the envelope group contained significantly more patients with ≥ 2 infection risk factors. Our results and those of other studies[9,12,24,26], support that the utilization of antibacterial envelopes (biologic or non-biologic) may reduce the potential risk burden of patients with multiple concurrent infection risk factors who are undergoing CIED procedures. However, further studies are needed to determine if there are specific patient types that could benefit the most from receiving an antibacterial envelope.

Complications

There were no significant differences in individual adverse event rates between groups, except that more patients with biologic envelopes were reported to have hematoma requiring intervention compared to the other two groups. However, this observation may have been due to the greater use of systemic anticoagulation and reoperative procedures in the biologic envelope group, which have both been shown to be risk factors for hematoma formation in previous studies[29,30]. In fact, a recent analysis of hematoma from the 6800 patients included in the WRAP-IT trial reported a hematoma occurrence of 2.2%, which was significantly associated with an increased risk of infection for the no envelope (control) group and a significantly lower risk of major infection in the non-biologic envelope group (2.5% vs 13.1%, P = 0.03)[31]. No hematoma in our dataset led to subsequent infection, which further supports a potential benefit from using antibacterial envelopes (biologic or non-biologic) to reduce the risk of hematoma manifesting to CIED implant site infection.

Infections at the CIED implantation site have serious morbidity, mortality, and economic consequences[1,32]. The use of antibacterial envelopes may reduce the risk of infection and could potentially reduce these serious complications and healthcare costs[33]. In our dataset, antibacterial envelopes were used significantly more often to treat patients with multiple comorbid risk factors, and biologic envelopes tended to be used more often in higher risk patients than non-biologic envelopes. We observed a 0.4% overall rate of pocket infection, which is lower than previously-reported studies of 0.7% to 4.6% for de novo implantations and up to 7% for reoperative procedures[5-8]. No significant difference was found in major CIED (pocket) infection rates between the 3 groups. A previous study reported that infection rate can differ depending upon various patient- and procedure-related circumstances (such as device type, procedure type, antibacterial envelope use, or perioperative antibiotics)[7], thus along with the major infection rates reported for high risk patients in the WRAP-IT (0.7%)[12] and PADIT (0.7%)[34] studies, the low pocket infection rate observed in our preliminary results (0.4%) supports that high infection risk factors can be countered with infection prophylaxis techniques such as the use of antibacterial envelopes.

Antibacterial CIED envelope types

There are currently two commercially available CIED envelopes in the United States. The biologic envelope (CanGaroo®) is manufactured from two sheets of 4-ply SIS-ECM material which can be hydrated by the implanting physician with an antibiotic solution prior to implantation, and the non-biologic envelope (TYRX™) is fabricated from an absorbable synthetic substrate mesh coated with a bioresorbable polyarylate polymer containing the drug substances rifampin and minocycline. In separate studies, the release of antibiotics occurs similarly from both envelopes over a period of seven days[10-13]. Both envelopes are intended to stabilize the CIED post-implantation, yet the host response to these different materials may vary. All biomaterials (biologic and non-biologic) interact with the body upon implantation, and certain characteristics of these materials can influence the host response to the implant[35,36].

Extensive studies have shown that implanted biologic materials (such as non-crosslinked decellularized SIS-ECM) stimulate the production of site appropriate, functional tissue (termed “constructive remodeling”[37])[20-23,36]. The ability to elicit a remodeling response post-implantation is due to the natural degradation of the implanted ECM by proteases which release intrinsic bioactive peptides and growth factors such as FGF-2 and VEGF in situ[22,38-40]. When implanted, for example into a CIED pocket, these bioavailable signaling molecules can influence the healing milieu surrounding the implant site by directing cellular activities such as differentiation, chemotaxis, adhesion, and angiogenesis[22,41-43]. Non-biologic materials do not contain these bioactive components.

Limitations

Limitations to this study include non-randomization of patients to the treatment groups, a limited period of follow up, and all implantations performed by a single physician at one institution. The choice of patients receiving an envelope (and which envelope was used) creates selection bias observed in the differing patient factors between groups. However, the intent of this report was to evaluate and define physician practice patterns instead of assessing superiority between the three therapies. Longer-term (> 1 year) follow up may have captured late adverse events, which cannot be ruled out in this study.

CONCLUSION

In this real-world study, patients at higher risk for CIED infection received antibacterial envelopes and lower infection risk patients did not receive envelopes, yet the CIED pocket infection rate did not differ between groups. There was also no significant difference in observed pocket infection rates for patients receiving biologic vs non-biologic antibacterial envelopes. These findings support that use of an antibacterial envelope may benefit patients who are at higher risk for infection, however further work will continue to refine patient selection and clinical decision-making for optimal utilization of antibacterial envelopes during CIED implantation.

ARTICLE HIGHLIGHTS
Research background

An increase in cardiac implantable electronic device (CIED) implantation has led to an increase in observed complication rates, including infection. Antibacterial CIED envelopes have been shown to reduce the risk of infection complications, even in high-risk patient groups. There are currently two different CIED envelopes in clinical use which differ in the material from which they are made.

Research motivation

There is a paucity of data describing real-world physician practice patterns when using antibacterial CIED envelopes. Understanding clinical rationale and outcomes from the use of this prophylactic therapy could improve future patient outcomes.

Research objectives

Patient risk profiles and outcomes were compared from patients undergoing CIED procedures receiving either no envelope, or one of two antibacterial envelopes.

Research methods

In this retrospective analysis, the records of consecutive CIED procedure patients were reviewed at one center through a follow-up time of 12 mo.

Research results

Patients who were selected to receive an antibacterial CIED envelope were at significantly higher risk for infection than patients who did not receive an envelope (81.2% vs 49.1%, P < 0.001). Among the infection risks, envelope patients were undergoing more reoperative procedures (47.1% vs 0.0%, P < 0.001) and received more high-powered devices (37.2% vs 5.8%, P = 0.004) than patients who received no envelope. There was a propensity for the physician choosing a biologic envelope in patients who were higher risk than non-biologic patients (84.3% vs 78.4%), and those that were undergoing reoperative procedures (62.9% vs 33.3%). The rate of pocket infection was low (any envelope 0.5% vs no envelope 0.0%), with no significant difference between the two envelope groups.

Research conclusions

There is an apparent benefit for using antibacterial envelopes in patients who are at higher risk of implant site infection. When using antibacterial envelopes, there was no significant difference in infection rate for biologic and non-biologic envelopes.

Research perspectives

Future studies should further explore patient and procedural factors that play a role in antibacterial envelope usage for specific patient types to further improve patient outcomes.

ACKNOWLEDGEMENTS

The authors acknowledge Sherrie Webb for writing and editing assistance and Kristina Chapple, PhD for her biostatistical analysis expertise.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author's Membership in Professional Societies: American Heart Association.

Specialty type: Cardiac and cardiovascular systems

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Apiratwarakul K, Thailand; Santomauro M, Italy S-Editor: Ma YJ L-Editor: A P-Editor: Ma YJ

References
1.  Baddour LM, Epstein AE, Erickson CC, Knight BP, Levison ME, Lockhart PB, Masoudi FA, Okum EJ, Wilson WR, Beerman LB, Bolger AF, Estes NA 3rd, Gewitz M, Newburger JW, Schron EB, Taubert KA; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee;  Council on Cardiovascular Disease in Young;  Council on Cardiovascular Surgery and Anesthesia;  Council on Cardiovascular Nursing;  Council on Clinical Cardiology;  Interdisciplinary Council on Quality of Care;  American Heart Association. Update on cardiovascular implantable electronic device infections and their management: a scientific statement from the American Heart Association. Circulation. 2010;121:458-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 743]  [Cited by in F6Publishing: 725]  [Article Influence: 51.8]  [Reference Citation Analysis (0)]
2.  Dai M, Cai C, Vaibhav V, Sohail MR, Hayes DL, Hodge DO, Tian Y, Asirvatham R, Cochuyt JJ, Huang C, Friedman PA, Cha YM. Trends of Cardiovascular Implantable Electronic Device Infection in 3 Decades: A Population-Based Study. JACC Clin Electrophysiol. 2019;5:1071-1080.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
3.  Greenspon AJ, Patel JD, Lau E, Ochoa JA, Frisch DR, Ho RT, Pavri BB, Kurtz SM. 16-year trends in the infection burden for pacemakers and implantable cardioverter-defibrillators in the United States 1993 to 2008. J Am Coll Cardiol. 2011;58:1001-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 493]  [Cited by in F6Publishing: 544]  [Article Influence: 41.8]  [Reference Citation Analysis (0)]
4.  Polyzos KA, Konstantelias AA, Falagas ME. Risk factors for cardiac implantable electronic device infection: a systematic review and meta-analysis. Europace. 2015;17:767-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in F6Publishing: 281]  [Article Influence: 31.2]  [Reference Citation Analysis (0)]
5.  Poole JE, Gleva MJ, Mela T, Chung MK, Uslan DZ, Borge R, Gottipaty V, Shinn T, Dan D, Feldman LA, Seide H, Winston SA, Gallagher JJ, Langberg JJ, Mitchell K, Holcomb R; REPLACE Registry Investigators. Complication rates associated with pacemaker or implantable cardioverter-defibrillator generator replacements and upgrade procedures: results from the REPLACE registry. Circulation. 2010;122:1553-1561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 560]  [Cited by in F6Publishing: 534]  [Article Influence: 38.1]  [Reference Citation Analysis (0)]
6.  Chung MK, Holcomb RG, Mittal S, Steinberg JS, Gleva MJ, Mela T, Uslan DZ, Mitchell K, Poole JE; REPLACE Investigators. REPLACE DARE (Death After Replacement Evaluation) score: determinants of all-cause mortality after implantable device replacement or upgrade from the REPLACE registry. Circ Arrhythm Electrophysiol. 2014;7:1048-1056.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
7.  Han HC, Hawkins NM, Pearman CM, Birnie DH, Krahn AD. Epidemiology of cardiac implantable electronic device infections: incidence and risk factors. Europace. 2021;23:iv3-iv10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 36]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
8.  Olsen T, Jørgensen OD, Nielsen JC, Thøgersen AM, Philbert BT, Johansen JB. Incidence of device-related infection in 97 750 patients: clinical data from the complete Danish device-cohort (1982-2018). Eur Heart J. 2019;40:1862-1869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 120]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
9.  Kolek MJ, Dresen WF, Wells QS, Ellis CR. Use of an antibacterial envelope is associated with reduced cardiac implantable electronic device infections in high-risk patients. Pacing Clin Electrophysiol. 2013;36:354-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 69]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
10.  Deering TF, Chang C, Snyder C, Natarajan SK, Matheny R. Enhanced Antimicrobial Effects of Decellularized Extracellular Matrix (CorMatrix) with Added Vancomycin and Gentamicin for Device Implant Protection. Pacing Clin Electrophysiol. 2017;40:615-623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
11.  Medtronic  Huntingdon Life Sciences Study TR-2013-001. 2013. Available from: URL: https://www.medtronic.com/us-en/healthcare-professionals/products/cardiac-rhythm/absorbable-antibacterial-envelopes/tyrx-envelope.html.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Tarakji KG, Mittal S, Kennergren C, Corey R, Poole JE, Schloss E, Gallastegui J, Pickett RA, Evonich R, Philippon F, McComb JM, Roark SF, Sorrentino D, Sholevar D, Cronin E, Berman B, Riggio D, Biffi M, Khan H, Silver MT, Collier J, Eldadah Z, Wright DJ, Lande JD, Lexcen DR, Cheng A, Wilkoff BL; WRAP-IT Investigators. Antibacterial Envelope to Prevent Cardiac Implantable Device Infection. N Engl J Med. 2019;380:1895-1905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 214]  [Article Influence: 42.8]  [Reference Citation Analysis (0)]
13.  Sohail MR, Esquer Garrigos Z, Elayi CS, Xiang K, Catanzaro JN. Preclinical evaluation of efficacy and pharmacokinetics of gentamicin containing extracellular-matrix envelope. Pacing Clin Electrophysiol. 2020;43:341-349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
14.  Holton LH 3rd, Chung T, Silverman RP, Haerian H, Goldberg NH, Burrows WM, Gobin A, Butler CE. Comparison of acellular dermal matrix and synthetic mesh for lateral chest wall reconstruction in a rabbit model. Plast Reconstr Surg. 2007;119:1238-1246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 40]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
15.  Laschke MW, Häufel JM, Scheuer C, Menger MD. Angiogenic and inflammatory host response to surgical meshes of different mesh architecture and polymer composition. J Biomed Mater Res B Appl Biomater. 2009;91:497-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
16.  Wolf MT, Carruthers CA, Dearth CL, Crapo PM, Huber A, Burnsed OA, Londono R, Johnson SA, Daly KA, Stahl EC, Freund JM, Medberry CJ, Carey LE, Nieponice A, Amoroso NJ, Badylak SF. Polypropylene surgical mesh coated with extracellular matrix mitigates the host foreign body response. J Biomed Mater Res A. 2014;102:234-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 89]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
17.  Lock AM, Gao R, Naot D, Coleman B, Cornish J, Musson DS. Induction of immune gene expression and inflammatory mediator release by commonly used surgical suture materials: an experimental in vitro study. Patient Saf Surg. 2017;11:16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 28]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
18.  Scislowska-Czarnecka A, Pamula E, Tlalka A, Kolaczkowska E. Effects of aliphatic polyesters on activation of the immune system: studies on macrophages. J Biomater Sci Polym Ed. 2012;23:715-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
19.  Parsonnet V. A stretch fabric pouch for implanted pacemakers. Arch Surg. 1972;105:654-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
20.  Londono R, Badylak SF. Biologic scaffolds for regenerative medicine: mechanisms of in vivo remodeling. Ann Biomed Eng. 2015;43:577-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 156]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
21.  Xiang K, Catanzaro JN, Elayi C, Esquer Garrigos Z, Sohail MR. Antibiotic-Eluting Envelopes to Prevent Cardiac-Implantable Electronic Device Infection: Past, Present, and Future. Cureus. 2021;13:e13088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
22.  Brown BN, Badylak SF. Extracellular matrix as an inductive scaffold for functional tissue reconstruction. Transl Res. 2014;163:268-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 310]  [Cited by in F6Publishing: 310]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
23.  Allen KB, Adams JD, Badylak SF, Garrett HE, Mouawad NJ, Oweida SW, Parikshak M, Sultan PK. Extracellular Matrix Patches for Endarterectomy Repair. Front Cardiovasc Med. 2021;8:631750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
24.  Nayak H, Beaser AD, Aziz ZA. Patient Profiles in the Utilization of the CanGaroo® Envelope. Cureus. 2021;13:e12702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
25.  Buchanan E, Yoo D. Use of Biologic Extracellular Matrix in Two Ways to Reduce Cardiac Electronic Device Infection. Cureus. 2021;13:e13037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
26.  Deering T. Antibiotic selection and risk profiles in patients receiving antibacterial cardiovascular implantable electronic device envelopes – A real world sample and analysis. Eur Heart J. 2021;42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
27.  Hercé B, Nazeyrollas P, Lesaffre F, Sandras R, Chabert JP, Martin A, Tassan-Mangina S, Bui HT, Metz D. Risk factors for infection of implantable cardiac devices: data from a registry of 2496 patients. Europace. 2013;15:66-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 56]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
28.  Lekkerkerker JC, van Nieuwkoop C, Trines SA, van der Bom JG, Bernards A, van de Velde ET, Bootsma M, Zeppenfeld K, Jukema JW, Borleffs JW, Schalij MJ, van Erven L. Risk factors and time delay associated with cardiac device infections: Leiden device registry. Heart. 2009;95:715-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 171]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
29.  Demir GG, Guler GB, Guler E, Güneş H, Kizilirmak F, Karaca İO, Omaygenç MO, Çakal B, Olgun E, Savur U, Ibisoglu E, Barutçu I, Kiliçaslan F. Pocket haematoma after cardiac electronic device implantation in patients receiving antiplatelet and anticoagulant treatment: a single-centre experience. Acta Cardiol. 2017;72:47-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
30.  Notaristefano F, Angeli F, Verdecchia P, Zingarini G, Spighi L, Annunziata R, Reccia MR, Piraccini S, Notaristefano S, Lip GYH, Cavallini C. Device-Pocket Hematoma After Cardiac Implantable Electronic Devices. Circ Arrhythm Electrophysiol. 2020;13:e008372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
31.  Tarakji KG, Korantzopoulos P, Philippon F, Biffi M, Mittal S, Poole JE, Kennergren C, Lexcen DR, Lande JD, Seshadri S, Wilkoff BL. Infectious consequences of hematoma from cardiac implantable electronic device procedures and the role of the antibiotic envelope: A WRAP-IT trial analysis. Heart Rhythm. 2021;18:2080-2086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
32.  Sohail MR, Eby EL, Ryan MP, Gunnarsson C, Wright LA, Greenspon AJ. Incidence, Treatment Intensity, and Incremental Annual Expenditures for Patients Experiencing a Cardiac Implantable Electronic Device Infection: Evidence From a Large US Payer Database 1-Year Post Implantation. Circ Arrhythm Electrophysiol. 2016;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
33.  Frausing MHJP, Kronborg MB, Johansen JB, Nielsen JC. Avoiding implant complications in cardiac implantable electronic devices: what works? Europace. 2021;23:163-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
34.  Krahn AD, Longtin Y, Philippon F, Birnie DH, Manlucu J, Angaran P, Rinne C, Coutu B, Low RA, Essebag V, Morillo C, Redfearn D, Toal S, Becker G, Degrâce M, Thibault B, Crystal E, Tung S, LeMaitre J, Sultan O, Bennett M, Bashir J, Ayala-Paredes F, Gervais P, Rioux L, Hemels MEW, Bouwels LHR, van Vlies B, Wang J, Exner DV, Dorian P, Parkash R, Alings M, Connolly SJ. Prevention of Arrhythmia Device Infection Trial: The PADIT Trial. J Am Coll Cardiol. 2018;72:3098-3109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 128]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
35.  Franz S, Rammelt S, Scharnweber D, Simon JC. Immune responses to implants - a review of the implications for the design of immunomodulatory biomaterials. Biomaterials. 2011;32:6692-6709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 918]  [Cited by in F6Publishing: 874]  [Article Influence: 67.2]  [Reference Citation Analysis (0)]
36.  Badylak SF. Decellularized allogeneic and xenogeneic tissue as a bioscaffold for regenerative medicine: factors that influence the host response. Ann Biomed Eng. 2014;42:1517-1527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 201]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
37.  Badylak SF, Brown BN, Gilbert TW, Daly KA, Huber A, Turner NJ. Biologic scaffolds for constructive tissue remodeling. Biomaterials. 2011;32:316-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
38.  Gilbert TW, Stewart-Akers AM, Simmons-Byrd A, Badylak SF. Degradation and remodeling of small intestinal submucosa in canine Achilles tendon repair. J Bone Joint Surg Am. 2007;89:621-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 166]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
39.  Reing JE, Brown BN, Daly KA, Freund JM, Gilbert TW, Hsiong SX, Huber A, Kullas KE, Tottey S, Wolf MT, Badylak SF. The effects of processing methods upon mechanical and biologic properties of porcine dermal extracellular matrix scaffolds. Biomaterials. 2010;31:8626-8633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 310]  [Cited by in F6Publishing: 306]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
40.  Swinehart IT, Badylak SF. Extracellular matrix bioscaffolds in tissue remodeling and morphogenesis. Dev Dyn. 2016;245:351-360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 133]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
41.  Li F, Li W, Johnson S, Ingram D, Yoder M, Badylak S. Low-molecular-weight peptides derived from extracellular matrix as chemoattractants for primary endothelial cells. Endothelium. 2004;11:199-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 126]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
42.  Davis GE. Matricryptic sites control tissue injury responses in the cardiovascular system: relationships to pattern recognition receptor regulated events. J Mol Cell Cardiol. 2010;48:454-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
43.  Brennan EP, Tang XH, Stewart-Akers AM, Gudas LJ, Badylak SF. Chemoattractant activity of degradation products of fetal and adult skin extracellular matrix for keratinocyte progenitor cells. J Tissue Eng Regen Med. 2008;2:491-498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 100]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]