Cardiac magnetic resonance imaging characterization of acute rejection in a porcine heterotopic heart transplantation model (2024)

  • Journal List
  • PLoS One
  • PMC11146723

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Cardiac magnetic resonance imaging characterization of acute rejection in a porcine heterotopic heart transplantation model (1)

Link to Publisher's site

PLoS One. 2024; 19(6): e0304588.

Published online 2024 Jun 3. doi:10.1371/journal.pone.0304588

PMCID: PMC11146723

PMID: 38829911

Michelle Mendiola Pla, Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Resources, Software, Validation, Visualization, Writing – original draft, Writing – review & editing,Cardiac magnetic resonance imaging characterization of acute rejection in a porcine heterotopic heart transplantation model (2)1,* Carmelo A. Milano, Conceptualization, Funding acquisition, Investigation, Project administration, Resources, Supervision, Writing – review & editing,1 Carolyn Glass, Data curation, Formal analysis, Visualization, Writing – review & editing,2 Dawn E. Bowles, Conceptualization, Formal analysis, Investigation, Methodology, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing,3 and David C. Wendell, Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Resources, Software, Validation, Visualization, Writing – original draft, Writing – review & editing4,5

Vincenzo Lionetti, Editor

Author information Article notes Copyright and License information PMC Disclaimer

Associated Data

Supplementary Materials
Data Availability Statement

Abstract

Preclinical disease models are important for the advancement of therapeutics towards human clinical trials. One of the difficult tasks of developing a well-characterized model is having a reliable modality with which to trend the progression of disease. Acute rejection is one of the most devastating complications that can occur following organ transplantation. Specifically in cardiac transplantation, approximately 12% of patients will experience at least one episode of moderate or severe acute rejection in the first year. Currently, the gold standard for monitoring rejection in the clinical setting is to perform serial endomyocardial biopsies for direct histological assessment. However, this is difficult to reproduce in a porcine model of acute rejection in cardiac transplantation where the heart is heterotopically transplanted in an abdominal position. Cardiac magnetic resonance imaging is arising as an alternative for serial screening for acute rejection in cardiac transplantation. This is an exploratory study to create and define a standardized cardiac magnetic resonance screening protocol for characterizing changes associated with the presence of acute rejection in this preclinical model of disease. Results demonstrate that increases in T1 mapping, T2 mapping, left ventricular mass, and in late gadolinium enhancement are significantly correlated with presence of acute rejection.

Introduction

Over 4,545 heart transplants were performed in 2023 for patients with advanced heart failure (accessed from https://optn.transplant.hrsa.gov/data/view-data-reports/national-data/). Over the last 30 years, the survival rate associated with transplantation has significantly improved [1]. However, acute rejection (AR) remains a major complication conferring substantial morbidity and mortality after transplantation. AR is among the most common complications within the first year after transplantation with 12.6% of recipients experiencing at least one episode of moderate or severe AR within the first year [2]. Incidence of even one episode of AR is a main determinant for increased morbidity post-transplantation.

Preclinical disease models provide a platform to replicate relevant clinical biology that characterizes human disease pathophysiology which can be used for the development of therapeutics. Model translatability ultimately depends on the fidelity observed between preclinical and clinical outcomes [3]. Hence, a crucial aspect of a reliable clinical model is to define methods and parameters to be able to measure the efficacy of an intervention. The porcine abdominal heterotopic heart transplantation model has been described as an ideal model for assessing the immunopathology of acute rejection. It is not reflective of typical cardiac physiology given the offloaded state of the heart as it does not receive a significant portion of blood volume from the recipient. However, it is advantageous over an orthotopic transplantation model due to better overall survival of the recipient. This is because the surgery for transplantation does not require cardiopulmonary bypass intra-operatively, and because there is no requirement of the allograft to maintain the recipient animal’s circulation [4, 5].

Currently, the gold standard for monitoring rejection in the clinical setting is to perform serial endomyocardial biopsies (EMB) for direct histological assessment. However, this is difficult to reproduce in a porcine model of acute rejection in cardiac transplantation where the heart is heterotopically transplanted into the abdomen of the animal. Cardiac magnetic resonance (CMR) imaging is arising as an alternative in the clinical setting for serial screening of acute rejection in cardiac transplantation. CMR has the advantage of being able to screen the entirety of the allograft myocardium, whereas EMB is only able to screen a small, random fraction of it. Evidence of increasing fibrosis, using late gadolinium enhancement, and edema, using T1 and T2 weighted mapping measurements, have been described to correlate with episodes of AR by several clinical studies, potentially making CMR more sensitive for the detection of AR than EMB [610].

There is currently no comprehensive or reliable way to monitor transplanted heterotopic abdominal cardiac grafts for AR. Studies that have used this preclinical model have described use of electrocardiography (ECG), cardiac pressure monitoring, myocardial tissue biopsy, blood marker monitoring, and palpation of the graft by physical exam have been described for monitoring for rejection [1113]. We describe the use CMR to screen transplanted abdominal hearts in a porcine model of acute rejection following heart transplantation and define the changes observed in cardiac activity and morphology using T1 mapping, T2 mapping, and late gadolinium enhancement measurements. We correlated these findings with results from adjunct EMB, blood marker monitoring, echocardiography, and palpation measurements.

Materials and methods

Cardiac transplantation and post-operative care

The care and treatment of pigs followed the Position of the American Heart Association on Research Animal Use [14]. This study was approved by the Duke University Institutional Animal Care and Use Committee (A122-22-06). Female Yucatan pigs (Sinclair Bio Resources, Auxvasse, MO) aged 7–9 months were used. The weight of the pigs ranged between 13-42kg. All pigs underwent SLA genotyping and blood-typing prior to selection. Pairs that were blood-type compatible and fully SLA Class 1 and Class 2 mismatched were utilized [4, 1517]. The pig weighing the least in each pair was selected for use as the donor animal. The donor hearts were procured in standard fashion and underwent normothermic ex vivo perfusion using a TransMedics Organ Care System device for 2 hours as described in Mendiola Pla et al. [4, 18]. The hearts were subsequently transplanted heterotopically into the recipient pig’s abdomen [4]. The study interventions and follow-up are outlined in Fig 1. For immunosuppression, the recipient pigs began receiving tacrolimus intramuscular injections three days prior to transplantation with a trough goal of 5-15ng/L. Following transplantation, the animals continued receiving tacrolimus injections. They were also started on a methylprednisolone taper and mycophenolate mofetil 400mg twice daily. The immunosuppression was stopped post-operative day (POD) 14 [19].

Open in a separate window

Fig 1

Study timeline of intervention and acute rejection follow-up.

Pig pairs underwent a “pre-op” CMR to measure baseline characteristics. Three days prior to abdominal heterotopic cardiac transplantation the pigs began immunosuppression therapy. The pigs then underwent transplantation on post-operative day 0. Immunosuppression therapy ended on post-operative day 14. “Rejection” CMR was performed on post-operative day 19 to assess for early onset of acute rejection in the allograft. Afterwards, transvenous endomyocardial biopsy was performed on post-operative day 23 and 30 in order to validate and trend the progression of acute rejection seen on CMR. After these procedures the cardiac allografts were monitored by physical exam until the allografts reached fulminant acute rejection as determined by all cessation of cardiac activity.

Animal health and behavior were monitored two to three times a day during the experimental period by trained personnel. The animals were sedated and underwent endotracheal mechanical ventilation for every invasive procedure and for CMR scans. General anesthesia using ketamine (5–33 mg/kg) and midazolam (0.2–0.5 mg/kg) intravenously was used then sedation was maintained using isoflurane gas (1–3%). Buprenorphine (0.005–0.01 mg/kg) was administered intramuscularly post-operatively for analgesia management. Cardiac activity was assessed daily by palpation and graded on a 0–4+ scale, where 0 corresponds to no palpable pulse whereas 4+ corresponds to a vigorous pulse that can be seen through the skin. Troponin blood levels and echocardiography were assessed weekly throughout the survival period.

Animals were survived until fulminant graft rejection was reached as determined by complete cessation of allograft activity on palpation and echocardiography or if allograft survival surpassed greater than 100 days. Once fulminant graft rejection was reached, animals were euthanized within 24 hours. If an allograft survived greater than 100 days, the animals were euthanized within one week of the timepoint. For euthanasia, animals again underwent endotracheal mechanical ventilation and sedation as described above. To procure the hearts, a sternotomy and laparotomy were performed and the hearts were each arrested using Del Nido cardioplegia infused directly into the respective aortic root of each heart. Each heart was subsequently excised from the animal.

Cardiac magnetic resonance

CMR studies were performed using a 3.0T MAGNETOM Vida scanner (Siemens Healthineers, Erlangen, Germany) equipped with a 32-channel chest coil and 72-channel spine array. CMR imaging was performed on both the donor and recipient animal up to 2 weeks prior to cardiac transplantation. Following transplantation another CMR imaging study was performed on POD 19 to characterize changes associated with AR. All pigs underwent sedation with general anesthesia and intubated for mechanical ventilatory support during the duration of the imaging studies. All images were acquired during ventilated breath holds and with cardiac gating applied using electrocardiography (ECG) [2022]. For the post-transplantation CMR studies, both the native and transplanted heart were able to be gated independently. To achieve this, one set of ECG electrode leads was arranged over the left chest to gate the animal’s native heart and another set was arranged over the right flank (Fig 2). The hearts were imaged sequentially with the ECG gating leads placed first on the chest when imaging the native heart then subsequently moved to the abdominal gating leads when imaging the transplanted heart.

Open in a separate window

Fig 2

Electrode lead and coil configuration.

Electrode lead configurations for obtaining the electrocardiogram measurements from the (A) recipient’s native heart in the thorax and from the (B) heterotopic cardiac allograft in the abdomen. (C) Two coils were used to obtain the CMR images from the native heart and the transplanted heart.

Different imaging techniques were obtained for analysis: Cine-CMR, T1 mapping, T2 mapping, and delayed enhancement CMR (DE-CMR). Cine-CMR was performed using a steady-state free-precession (SSFP) sequence with short-axis images acquired from the level of the mitral valve annulus though the apex (slice thickness 6mm; no gap; temporal resolution: <35ms). T1 mapping images were acquired using 5(3)3 sampling algorithm at slice locations corresponding to cine images. Spatial resolution was 1.0cm x 1.0cm, slice thickness 6mm, and temporal resolution <150ms. T1 mapping raw images were inspected for any mis-registered images, and if necessary, offline T1 mapping analysis was performed. T2 mapping images were also acquired at the same slice locations as cine and T1 mapping images. This sequence consisted of 3 single-shot SSFP images with increasing T2 preparation times (0ms, 25ms, 55ms).

DE-CMR images were obtained 15-minutes after administering gadoterate meglumine (0.15mmol/kg) intravenously. DE-CMR images were obtained using the following parameters: slice thickness: 6mm; in-plane spatial resolution: 1.0mm ×1.0mm; temporal resolution: 180ms; trigger pulse: 2 R-R intervals; breath hold time: 10-15ms. A segmented, inversion-recovery gradient-echo sequence with magnitude and phase-sensitive reconstructions were used for DE-CMR, with inversion time manually selected to null normal myocardium. Finally, velocity encoded images were acquired near the anastomotic sites for the donor heart to verify patent flow to the graft.

Analysis of cardiac magnetic resonance images

Pre- and post-transplantation measurements obtained from the CMR scans were compared for both the native heart and the donor heart. Cine-CMR images were used to calculate ejection fraction (EF) and left ventricular (LV) mass. T1 and T2 mapping values were quantified according to the Society of Cardiovascular Magnetic Resonance guidelines, avoiding the most subendocardial and subepicardial portions of the heart [23]. DE-CMR images were analyzed quantitatively using a full-width-half-max method [24], and the amount of fibrosis/necrosis was compared with baseline image findings.

Tissue biopsy collection

Animals were sedated using general anesthesia and intubated for ventilation. For endomyocardial biopsy, the procedure was performed as described in Mendiola Pla et al. on POD 23 and 30 [25]. Briefly, an 8-Fr introducer sheath was inserted into the femoral vein and a 7-Fr bioptome was inserted through the sheath and fluoroscopically guided into the right ventricle of the cardiac allograft to obtain the biopsies. Sharp biopsies were obtained at the time of fulminant allograft rejection at which time the animal was euthanized and the allograft was excised.

Histology and rejection grading

All biopsies were fixed in 10% neutral buffered formalin prior to paraffin embedding. 5-micron thick sections were prepared and stained with hematoxylin and eosin (H&E) to assess for rejection grade or with Masson’s trichrome stain to assess for fibrosis. Sections were assessed and graded according to ISHLT guidelines by a cardiac pathologist, CG, who was blinded to the CMR results [26].

Statistics

Statistical analysis was performed using GraphPad Prism 10 Version 10.1.0 (316) (GraphPad Software, Inc. La Jolla, CA). A p-value of <0.05 was considered significant. Normality was assessed using Shapiro-Wilk test. For comparisons between pre-transplantation and post-transplantation measurements with parametric distributions, the changes in mean values of each group were calculated and compared using Student’s t-test. For those with non-parametric distribution, Mann Whitney U test was used. Statistical notations used in the figures: p>0.05, not significant (ns); p<0.05, *; p<0.01, **; p<0.001, ***; p<0.0001, ****.

Results

Each animal (n = 14) underwent a baseline CMR scan and each transplant recipient animal (n = 6) underwent a rejection assessment CMR scan on POD 19. All CMR scans were able to be fully completed on all animals without any complications. One transplant recipient animal died in the immediate post-operative period from surgical complications related to bleeding and was excluded from analysis. Of the n = 6 transplanted hearts, n = 5 progressed to fulminant rejection; n = 1 did not progress to fulminant AR by POD 106 and was euthanized at that time. The average time to fulminant rejection following transplantation was 51±22 days (mean±SD) (n = 5).

Baseline CMR

Baseline CMR measurements are listed in Table 1. Native hearts had an average mass of 57.4±9.0g and allograft heart had a mass of 50.3±8.1g (mean±SD). The average ejection fraction for native hearts was 69.3±7.6% and for allograft hearts was 62.5±7.0% (mean±SD). T1 mapping values were 1138±8.9ms for native hearts and 1144±28.7ms for allograft hearts (p = 0.69). T2 mapping values were 36.9±1.3ms for native hearts and 36.8±1.8mg for allograft hearts (p = 0.9). Collectively, the T1 and T2 measurements indicate that there was no evidence of inflammation or edema of the myocardium prior to transplantation. DE-CMR measurements were negative for fibrosis in both native and donor hearts prior to transplantation.

Table 1

Baseline CMR characteristics.

Recipient (n = 6)Allograft
(n = 6)
LV Mass, g (mean±SD)57.4±9.050.3±8.1
Ejection fraction, % (mean±SD)69.3±7.662.5±7.0
T1 mapping, ms1138±8.91144±28.7
T2 mapping, ms36.9±1.336.8±1.8
Delayed enhancement, %00

Open in a separate window

Post-transplantation CMR in the setting of rejection

Post-transplantation CMR results are listed in Table 2 and Fig 3. At 19-days post-transplantation there was significant increase in LV mass in the allograft heart, 63.5±16.8g, compared to the native heart, 8.3±2.5g (p<0.0001). There was also a significant increase in T1 mapping values from baseline in the allograft heart (222.6±55.0ms) relative to the native heart (77.8±41.9ms) (p = 0.002). The increase in T2 mapping values was 6.0±3.1ms for native hearts and 13.7±4.2ms for allograft hearts (p = 0.008). DE-CMR mean percent area of enhancement was 6.5% in the allograft hearts versus 0% in the native hearts (p = 0.008). Velocity measurements across the aortic anastomoses did not show any flow acceleration, demonstrating no areas of inflow stenosis.

Open in a separate window

Fig 3

Cardiac magnetic resonance imaging results.

Representative CMR T1 mapping, T2 mapping, and DE-CMR images (Animal D) of the native and donor hearts on post-operative day 19. There was marked T1 elevation, T2 elevation, increase in LV mass, and fibrosis as noted by increased DE-CMR percentage. Cumulative results were plotted from ΔT1 measurements, ΔT2 measurements, ΔLV mass, and DE-CMR percentage. (*) <0.05, (**) <0.001, (***) <0.0001.

Table 2

Post-transplantation CMR acute rejection characteristics.

Native
(n = 5)
Allograft
(n = 5)
p-value
ΔLV Mass, g (mean±SD)8.3±2.563.5±16.8<0.0001
ΔT1 mapping, ms77.8±41.9222.6±55.00.002
ΔT2 mapping, ms6.0±3.113.7±4.20.008
Delayed enhancement, %06.3±6.20.008

Open in a separate window

Post-transplantation CMR in the setting of no rejection

In the animal that did not develop fulminant rejection within the study timeframe, CMR demonstrated an increase of 46.2g in allograft heart LV mass relative to just 10.8g in the native heart. Change in T1 mapping values were in the allograft heart (101.6ms) relative to the native heart (65.5ms). Change in T2 mapping values were slightly increased (9.9ms) in the allograft heart versus (3.1ms) in the native heart. However, both measurements did not reach the levels observed in the animals that did progress to fulminant rejection. Interestingly, DE-CMR demonstrated diffuse and scattered myocardial fibrosis (9.7%) despite other measures of AR being less marked. At the time of donor heart explantation, gross inspection demonstrated small multifocal infarcts scattered throughout the myocardium suggestive of a shower embolic event accounting for this finding on CMR and pathology.

Supplemental measures of acute rejection

EMB on POD 19 was successfully collected from n = 4 allografts and from n = 3 on POD 30. H&E at POD 19 demonstrated evidence of rejection on n = 2 samples, however no rejection was noted in n = 2 samples (Fig 4A). Of note, one of these samples originated from the allograft that did not reach fulminant rejection prior to the end of study. H&E at POD 30 demonstrated AR on all n = 3 samples obtained. Masson’s trichrome stain on EMBs obtained at POD 19 and POD 30 demonstrated fibrosis in all of the samples (Fig 4B). Average troponin measurements at baseline (221.5±210.7ng/L) were lower than the troponin measurements on POD 19 (7764.4±10398.4ng/L) (mean±SD). At POD 19 the median palpation grade was 3 with an interquartile range of 3 to 3.75. All of the individual supplemental measures are presented in Table 3.

Open in a separate window

Fig 4

Representative histopathology results of acute rejection.

Representative histopathology results of the hearts from one of the transplanted animals (Animal D). Endomyocardial biopsy samples from post-operative day 23 stained for (A) fibrosis (Masson Trichrome) and (B) lymphocytic infiltration (H&E). Both biopsy samples were consistent with acute rejection. At the time of euthanasia on post-operative day 54, the (C) native heart did not show evidence inflammation or scarring, whereas the (D) allograft heart had diffuse inflammation and scarring throughout.

Table 3

Correlating measures of allograft injury and rejection.

AnimalRejection POD 23Rejection POD 30Fibrosis POD 23Fibrosis POD 30Troponin (ng/L)Palpation gradeTime to fulminant rejection (days)
A----434341
B----1458489
CNo rejectionAcute rejectionFibrosis-11185333
DAcute rejectionAcute rejectionFibrosisFibrosis1144353
EAcute rejectionAcute rejectionFibrosisFibrosis24601439
FNo rejection-Fibrosis-5263n/a

Open in a separate window

Discussion

This exploratory study presents the utility of CMR in characterizing AR in a porcine preclinical model of AR in the context of cardiac transplantation, providing comprehensive data for screening for AR [19]. It demonstrates that this modality for examining the allograft is reliable for observing changes in an intra-abdominally transplanted cardiac allograft. This preclinical model of heterotopic heart transplantation is versatile for examining the immunological changes mounted by the recipient and morphological changes that the allograft undergoes at advanced stages of acute rejection [27]. The main limitation of the model across all of these studies is that there is not a highly sensitive method with low associated morbidity that is able to monitor for rejection in the allograft [4, 12, 13, 28]. We used CMR to characterize AR of the allograft on POD 19 after transplantation. Change in LV mass was significantly greater in the donor allograft compared to native hearts. The small increase in mass that is noted in the native hearts is attributed to the expected growth of the pig over the interval period between the baseline CMR and POD 19 CMR. Both T1 and T2 mapping values were each significantly elevated in the donor hearts relative to the native hearts. DE-CMR also demonstrated an increase in fibrotic changes within rejected myocardium that was significant.

The current clinical gold standard for monitoring AR is through EMB. EMB is an invasive procedure that can result in serious complications that compromise the health of the individual. CMR is being used more widely as an alternative diagnostic modality to monitor cardiac allografts post-transplantation. Advances in magnetic resonance imaging technology have allowed for detailed tissue characterization of the heart to be obtained non-invasively with no morbidity associated with the procedure. Many CMR protocols for AR monitoring have been described in the literature and validated against EMB-confirmed incidences of AR. Most clinical studies describe that T2 mapping and ECV values correlate significantly with of AR and while T1 mapping values tended to trend higher they never reached statistical significance in these studies [7, 8, 10]. One study observed that T1 mapping values correlated strongly with episodes of AR and demonstrated correction to baseline values after immunosuppressive therapy [9]. Anthony et al. observed that T1 mapping values and T2 mapping values individually correlated with AR, however when combined in a multiparametric fashion the diagnostic sensitivity was higher than either value alone [6]. As described above, our findings agree with the conclusions of these studies that T1 and T2 mapping values become elevated with the presence of AR.

Several techniques have been described previously for monitoring donor graft rejection in this preclinical model. Transabdominal echocardiography is easy to perform where the contractility of the ventricles, LV hypertrophy, and presence of LV thrombus can be assessed. However, consistency of measurements is subject to user variability. Obtaining transabdominal biopsies of the allograft has been used by several groups to be able to monitor the donor graft over multiple timepoints [12, 13]. The transabdominal biopsy technique is limited to one time in these studies since it involves a high risk of injuring the surrounding small bowel or causing significant hemorrhage from the allograft. One group has described the implantation of a telemetry device to measure atrial and ventricular pressures as well as ECG recordings to monitor for rejection [13]. They observed progressive LV hypertrophy and the presence of thrombus on ultrasound in conjunction with increasing troponin and thrombocytopenia, and most definitively an increase in LV end diastolic pressure. However, the technology and environmental setup to be able to use an implantable telemetry device is very expensive and requires a specially outfitted room in order to obtain the recordings. Another group described using daily palpation of the graft with ECG performed twice a week to detect when the heart rate went below 60 beats per minute. Echocardiography was then used to assess systolic function of the graft. Each of these episodes was considered an episode of AR [28]. While this group described this technique to correlate well with AR, this is an indirect method and episodes of bradycardia without continuous monitoring could readily be missed during early onset of AR. Addition of CMR for monitoring the allograft in this model confers advantages, such as the ability to assess the morphology of the heart in a completely non-invasive manner where both the donor and the native heart can be analyzed at the same time.

Serial CMR scans were not able to be performed in each animal, which is a limitation of this exploratory study. As a result, we were unable to characterize the gradual progression of rejection in the allografts towards fulminant rejection within this preclinical model. Future studies utilizing CMR for screening of AR rejection in this preclinical model would be needed to elucidate changes beyond the scope of this proof-of-concept study. However, given the other adjuncts described to monitor rejection, by POD 19 we were able to observe significant changes in the allografts consistent with changes in rejection.

Conclusion

Application of CMR for monitoring of acute rejection in a porcine model of acute rejection is a safe modality that offers the ability to monitor the entirety of the cardiac allograft and to quantify changes in its functionality. CMR in this setting offers a comprehensive and consistent method for AR monitoring.

Supporting information

S1 Data

(XLSX)

Click here to view.(23K, xlsx)

S1 Video

Allograft CMR.

Pre-op cine clip of the allograft heart followed by the post-op cine clip of the allograft heart.

(3GP)

Click here to view.(229K, 3gp)

S2 Video

Native CMR.

Pre-op cine clip of the recipient’s native heart followed by the post-op cine clip of the native heart.

(3GP)

Click here to view.(183K, 3gp)

Acknowledgments

We thank the members of the Duke Division of Laboratory Animal Resources for supporting the animals during the MRI procedures and the Duke Cardiovascular Magnetic Resonance Center for providing use of their facility to perform the CMR studies. We thank Sam Ho and Lynden Gault for performing the genotyping of the animals used in this study. We also thank Amy Evans and Duke Perfusion Services for providing perfusion support during each transplant surgery.

Funding Statement

CAM received funding from the Duke Division of Cardiothoracic Surgery and from TransMedics, Inc. The funders did not play a role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

References

1. Bakhtiyar SS, Godfrey EL, Ahmed S, Lamba H, Morgan J, Loor G, et al. Survival on the Heart Transplant Waiting List.JAMA Cardiology. 2020;5(11):1227–35. doi: 10.1001/jamacardio.2020.2795 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

2. Khush KK, Cherikh WS, Chambers DC, Harhay MO, Hayes D Jr, Hsich E, et al. The International Thoracic Organ Transplant Registry of the International Society for Heart and Lung Transplantation: Thirty-sixth adult heart transplantation report—2019; focus theme: Donor and recipient size match.J Heart Lung Transplant.2019;38(10):1056–66. Epub 20190810. doi: 10.1016/j.healun.2019.08.004 ; PubMed Central PMCID: PMC6816343. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

3. Honkala A, Malhotra SV, Kummar S, Junttila MR. Harnessing the predictive power of preclinical models for oncology drug development. Nature Reviews Drug Discovery. 2022;21(2):99–114. doi: 10.1038/s41573-021-00301-6 [PubMed] [CrossRef] [Google Scholar]

4. Mendiola Pla M, Evans A, Lee FH, Chiang Y, Bishawi M, Vekstein A, et al. A Porcine Heterotopic Heart Transplantation Protocol for Delivery of Therapeutics to a Cardiac Allograft.JoVE. 2022;(180):e63114. doi: 10.3791/63114 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

5. Kadner A, Chen RH, Adams DH. Heterotopic heart transplantation: experimental development and clinical experience. Eur J Cardiothorac Surg. 2000;17(4):474–81. doi: 10.1016/s1010-7940(00)00362-6 . [PubMed] [CrossRef] [Google Scholar]

6. Anthony C, Imran M, Pouliopoulos J, Emmanuel S, Iliff J, Liu Z, et al. Cardiovascular Magnetic Resonance for Rejection Surveillance After Cardiac Transplantation. Circulation. 2022;145(25):1811–24. doi: 10.1161/CIRCULATIONAHA.121.057006 [PubMed] [CrossRef] [Google Scholar]

7. Soslow JH, Godown J, Bearl DW, Crum K, Slaughter JC, George-Durrett K, et al. Cardiac Magnetic Resonance Imaging Noninvasively Detects Rejection in Pediatric Heart Transplant Recipients. Circulation: Cardiovascular Imaging.2022;15(4):e013456. doi: 10.1161/CIRCIMAGING.121.013456 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

8. Vermes E, Pantaléon C, Auvet A, Cazeneuve N, Machet MC, Delhommais A, et al. Cardiovascular magnetic resonance in heart transplant patients: diagnostic value of quantitative tissue markers: T2 mapping and extracellular volume fraction, for acute rejection diagnosis.J Cardiovasc Magn Reson.2018;20(1):59. Epub 20180827. doi: 10.1186/s12968-018-0480-9 ; PubMed Central PMCID: PMC6114788. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

9. Imran M, Wang L, McCrohon J, Yu C, Holloway C, Otton J, et al. Native T1 Mapping in the Diagnosis of Cardiac Allograft Rejection: A Prospective Histologically Validated Study.JACC: Cardiovascular Imaging.2019;12(8, Part 2):1618–28. doi: 10.1016/j.jcmg.2018.10.027 [PubMed] [CrossRef] [Google Scholar]

10. Dolan RS, Rahsepar AA, Blaisdell J, Suwa K, Ghafourian K, Wilcox JE, et al. Multiparametric Cardiac Magnetic Resonance Imaging Can Detect Acute Cardiac Allograft Rejection After Heart Transplantation.JACC Cardiovasc Imaging. 2019;12(8 Pt 2):1632–41. Epub 20190313. doi: 10.1016/j.jcmg.2019.01.026 ; PubMed Central PMCID: PMC6995349. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

11. Mendiola Pla M, Chiang Y, Roki A, Wang C, Lee FH, Smith MF, et al. Ex Vivo Gene Delivery to Porcine Cardiac Allografts Using a Myocardial-Enhanced Adeno-Associated Viral Vector. Hum Gene Ther. 2023;34(7–8):303–13. doi: 10.1089/hum.2022.241 ; PubMed Central PMCID: PMC10325812. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

12. Madsen JC, Sachs DH, Fallon JT, Weissman NJ. Cardiac allograft vasculopathy in partially inbred miniature swine. I. Time course, pathology, and dependence on immune mechanisms. The Journal of Thoracic and Cardiovascular Surgery. 1996;111(6):1230–9. doi: 10.1016/s0022-5223(96)70226-x [PubMed] [CrossRef] [Google Scholar]

13. Goerlich CE, DiChiacchio L, Zhang T, Singh AK, Lewis B, Tatarov I, et al. Heterotopic Porcine Cardiac Xenotransplantation in the Intra-Abdominal Position in a Non-Human Primate Model.Scientific Reports. 2020;10(1):10709. doi: 10.1038/s41598-020-66430-x [PMC free article] [PubMed] [CrossRef] [Google Scholar]

14. Position of the American Heart Association on research animal use.Circulation. 1985;71(4):849a–50a. . [PubMed] [Google Scholar]

15. Smith DM, Martens GW, Ho CS, Asbury JM. DNA sequence based typing of swine leukocyte antigens in Yucatan miniature pigs. Xenotransplantation. 2005;12(6):481–8. doi: 10.1111/j.1399-3089.2005.00252.x . [PubMed] [CrossRef] [Google Scholar]

16. Ho CS, Lunney JK, Franzo-Romain MH, Martens GW, Lee YJ, Lee JH, et al. Molecular characterization of swine leucocyte antigen class I genes in outbred pig populations. Anim Genet. 2009;40(4):468–78. Epub 20090320. doi: 10.1111/j.1365-2052.2009.01860.x . [PubMed] [CrossRef] [Google Scholar]

17. Ho CS, Lunney JK, Lee JH, Franzo-Romain MH, Martens GW, Rowland RR, et al. Molecular characterization of swine leucocyte antigen class II genes in outbred pig populations. Anim Genet. 2010;41(4):428–32. Epub 20100131. doi: 10.1111/j.1365-2052.2010.02019.x . [PubMed] [CrossRef] [Google Scholar]

18. Mendiola Pla M, Evans A, Lezberg P, Bowles DE. Ex Vivo Delivery of Viral Vectors by Organ Perfusion for Cardiac Transplantation Gene Therapy. In: Ishikawa K, editor. Cardiac Gene Therapy: Methods and Protocols.New York, NY: Springer US; 2022. p. 249–59. [PubMed] [Google Scholar]

19. Mendiola Pla M, Chiang Y, Glass C, Wendell D, Lenz DS, Kang L, et al. (987) A Porcine Model of Acute Rejection for Cardiac Transplantation.The Journal of Heart and Lung Transplantation. 2023;42(4, Supplement):S425. 10.1016/j.healun.2023.02.1098. [CrossRef] [Google Scholar]

20. Kim HW, Van Assche L, Jennings RB, Wince WB, Jensen CJ, Rehwald WG, et al. Relationship of T2-Weighted MRI Myocardial Hyperintensity and the Ischemic Area-At-Risk. Circ Res. 2015;117(3):254–65. Epub 20150513. doi: 10.1161/CIRCRESAHA.117.305771 ; PubMed Central PMCID: PMC4503326. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

21. Kim HW, Rehwald WG, Jenista ER, Wendell DC, Filev P, van Assche L, et al. Dark-Blood Delayed Enhancement Cardiac Magnetic Resonance of Myocardial Infarction.JACC Cardiovasc Imaging.2018;11(12):1758–69. Epub 20171213. doi: 10.1016/j.jcmg.2017.09.021 ; PubMed Central PMCID: PMC5993564. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

22. Wendell D, Jenista E, Kim HW, Chen EL, Azevedo CF, Kaolawanich Y, et al. Assessment of Papillary Muscle Infarction with Dark-Blood Delayed Enhancement Cardiac MRI in Canines and Humans.Radiology. 2022;305(2):329–38. Epub 20220726. doi: 10.1148/radiol.220251 ; PubMed Central PMCID: PMC9619201. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

23. Schulz-Menger J, Bluemke DA, Bremerich J, Flamm SD, Fogel MA, Friedrich MG, et al. Standardized image interpretation and post-processing in cardiovascular magnetic resonance—2020 update: Society for Cardiovascular Magnetic Resonance (SCMR): Board of Trustees Task Force on Standardized Post-Processing.J Cardiovasc Magn Reson.2020;22(1):19. Epub 20200312. doi: 10.1186/s12968-020-00610-6 ; PubMed Central PMCID: PMC7066763. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

24. Flett AS, Hasleton J, Cook C, Hausenloy D, Quarta G, Ariti C, et al. Evaluation of techniques for the quantification of myocardial scar of differing etiology using cardiac magnetic resonance.JACC Cardiovasc Imaging. 2011;4(2):150–6. doi: 10.1016/j.jcmg.2010.11.015 . [PubMed] [CrossRef] [Google Scholar]

25. Mendiola Pla M, Milano CA, Chiang Y, Bishawi M, Kang L, Lee FH, et al. Transvenous Endomyocardial Biopsy Technique for Intra-abdominal Heterotopic Cardiac Grafts.J Cardiovasc Transl Res.2023;16(3):748–50. Epub 20221107. doi: 10.1007/s12265-022-10337-7 ; PubMed Central PMCID: PMC10201646. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

26. Stewart S, Winters GL, Fishbein MC, Tazelaar HD, Kobashigawa J, Abrams J, et al. Revision of the 1990 working formulation for the standardization of nomenclature in the diagnosis of heart rejection. J Heart Lung Transplant. 2005;24(11):1710–20. Epub 20050620. doi: 10.1016/j.healun.2005.03.019 . [PubMed] [CrossRef] [Google Scholar]

27. Mendiola Pla M, Chiang Y, Roan J-N, E. Bowles D. Gene Therapy for Cardiac Transplantation. IntechOpen; 2022. [Google Scholar]

28. Hsu WT, Lin CH, Jui HY, Tseng YH, Shun CT, Hsu MC, et al. CXCR4 Antagonist Reduced the Incidence of Acute Rejection and Controlled Cardiac Allograft Vasculopathy in a Swine Heart Transplant Model Receiving a Mycophenolate-based Immunosuppressive Regimen. Transplantation. 2018;102(12):2002–11. doi: 10.1097/TP.0000000000002404 ; PubMed Central PMCID: PMC6257103. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Articles from PLOS ONE are provided here courtesy of PLOS

Cardiac magnetic resonance imaging characterization of acute rejection in a porcine heterotopic heart transplantation model (2024)
Top Articles
Latest Posts
Article information

Author: Velia Krajcik

Last Updated:

Views: 5838

Rating: 4.3 / 5 (74 voted)

Reviews: 89% of readers found this page helpful

Author information

Name: Velia Krajcik

Birthday: 1996-07-27

Address: 520 Balistreri Mount, South Armand, OR 60528

Phone: +466880739437

Job: Future Retail Associate

Hobby: Polo, Scouting, Worldbuilding, Cosplaying, Photography, Rowing, Nordic skating

Introduction: My name is Velia Krajcik, I am a handsome, clean, lucky, gleaming, magnificent, proud, glorious person who loves writing and wants to share my knowledge and understanding with you.