1
|
Inglis SS, Abbas M, Asleh R, Garmany A, Smith BH, Kushwaha S, Pereira N, Clavell AL, Villavicencio MA, Spencer PJ, Daly RC, Behfar A, Rosenbaum AN. Incidence and risk factors for rejection after conversion from calcineurin inhibitor to sirolimus-based immunosuppression in orthotopic heart transplant recipients. J Heart Lung Transplant 2025; 44:975-983. [PMID: 39743050 DOI: 10.1016/j.healun.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Although recommended in International Society for Heart and Lung Transplantation (ISHLT) guidelines, transition to mammalian targets of rapamycin (mTOR) inhibitors in heart transplant recipients is not routinely performed, in part due to perceived risk of rejection. This study sought to evaluate the incidence and risk factors for biopsy-proven, clinically relevant rejection following conversion from calcineurin inhibitor (CNI) to sirolimus (SRL) immunosuppression. METHODS A single center retrospective study was conducted of all consecutive adult patients who underwent orthotopic heart transplantation (OHT) and CNI-free SRL conversion from January 1999 to January 2023. All post-OHT biopsy data were obtained and graded per ISHLT criteria (antibody-mediated rejection [pAMR] or acute cellular rejection [ACR]). The primary endpoint was early rejection, defined as grade 2R ACR, pAMR 1, or greater, within 6 months after conversion. RESULTS Three hundred and seventeen patients (72% male, mean age 51.5±12.6 years) were included. Median time to SRL conversion following OHT was 0.76 years (IQR 0.49, 1.42). Median time from conversion to rejection was 0.51 years (IQR 0.31, 1.05). Thirty eight patients (12%) experienced early rejection. Following multivariate analysis, both timing to SRL conversion following OHT (OR 0.94 per month, 95% CI: 0.89-0.99, p-value=0.0054) and age at transplantation (OR 0.96, 95% CI: 0.93-0.99, p-value=0.0071) were independently associated with early rejection. Rejection following SRL conversion was not associated with increased risk of cardiac allograft vasculopathy (CAV) grade 2-3. CONCLUSIONS In a CNI-free SRL conversion protocol, both earlier SRL conversion following OHT and younger age at transplant are independently associated with early rejection, but rejection is not associated with a net increased risk of prognostically important CAV. Individualization of transition is necessary to mitigate risk, and these findings may aid in improvement of future conversion protocols.
Collapse
Affiliation(s)
- Sara S Inglis
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota; Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mohsin Abbas
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota; Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Rabea Asleh
- Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Armin Garmany
- Graduate School of Biomedical Sciences, Alix School of Medicine, Medical Scientist Training Program, Mayo Clinic, Rochester, Minnesota
| | - Byron H Smith
- Department of Biostatistics, Mayo Clinic, Rochester, Minnesota
| | - Sudhir Kushwaha
- Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Naveen Pereira
- Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alfredo L Clavell
- Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Philip J Spencer
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | - Richard C Daly
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, Minnesota
| | - Atta Behfar
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota; Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Andrew N Rosenbaum
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota; Deparment of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
2
|
Thorp EB, Ananthakrishnan A, Lantz CW. Decoding immune cell interactions during cardiac allograft vasculopathy: insights derived from bioinformatic strategies. Front Cardiovasc Med 2025; 12:1568528. [PMID: 40342971 PMCID: PMC12058854 DOI: 10.3389/fcvm.2025.1568528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025] Open
Abstract
Chronic allograft vasculopathy (CAV) is a major cause of late graft failure in heart transplant recipients, characterized by progressive intimal thickening and diffuse narrowing of the coronary arteries. Unlike atherosclerosis, CAV exhibits a distinct cellular composition and lesion distribution, yet its pathogenesis remains incompletely understood. A major challenge in CAV research has been the limited application of advanced "-omics" technologies, which have revolutionized the study of other vascular diseases. Recent advancements in single-cell and spatial transcriptomics, proteomics, and metabolomics have begun to uncover the complex immune-endothelial-stromal interactions driving CAV progression. Notably, single-cell RNA sequencing has identified previously unrecognized immune cell populations and signaling pathways implicated in endothelial injury and vascular remodeling after heart transplantation. Despite these breakthroughs, studies applying these technologies to CAV remain sparse, limiting the translation of these insights into clinical practice. This review aims to bridge this gap by summarizing recent findings from single-cell and multi-omic approaches, highlighting key discoveries, and discussing their implications for understanding CAV pathogenesis.
Collapse
Affiliation(s)
- Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Aparnaa Ananthakrishnan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Connor W. Lantz
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
3
|
Amancherla K, Taravella Oill AM, Bledsoe X, Williams AL, Chow N, Zhao S, Sheng Q, Bearl DW, Hoffman RD, Menachem JN, Siddiqi HK, Brinkley DM, Mee ED, Hadad N, Agrawal V, Schmeckpepper J, Rali AS, Tsai S, Farber-Eger EH, Wells QS, Freedman JE, Tucker NR, Schlendorf KH, Gamazon ER, Shah RV, Banovich N. Dynamic responses to rejection in the transplanted human heart revealed through spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640852. [PMID: 40093136 PMCID: PMC11908199 DOI: 10.1101/2025.02.28.640852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Allograft rejection following solid-organ transplantation is a major cause of graft dysfunction and mortality. Current approaches to diagnosis rely on histology, which exhibits wide diagnostic variability and lacks access to molecular phenotypes that may stratify therapeutic response. Here, we leverage image-based spatial transcriptomics at sub-cellular resolution in longitudinal human cardiac biopsies to characterize transcriptional heterogeneity in 62 adult and pediatric heart transplant (HT) recipients during and following histologically-diagnosed rejection. Across 28 cell types, we identified significant differences in abundance in CD4 + and CD8 + T cells, fibroblasts, and endothelial cells across different biological classes of rejection (cellular, mixed, antibody-mediated). We observed a broad overlap in cellular transcriptional states across histologic rejection severity and biological class and significant heterogeneity within rejection severity grades that would qualify for immunomodulatory treatment. Individuals who had resolved rejection after therapy had a distinct transcriptomic profile relative to those with persistent rejection, including 216 genes across 6 cell types along pathways of inflammation, IL6-JAK-STAT3 signaling, IFNα/IFNγ response, and TNFα signaling. Spatial transcriptomics also identified genes linked to long-term prognostic outcomes post-HT. These results underscore importance of subtyping immunologic states during rejection to stratify immune-cardiac interactions following HT that are therapeutically relevant to short- and long-term rejection-related outcomes.
Collapse
|
4
|
Jain R, Kransdorf EP, Cowger J, Jeevanandam V, Kobashigawa JA. Donor Selection for Heart Transplantation in 2025. JACC. HEART FAILURE 2025; 13:389-401. [PMID: 39570235 DOI: 10.1016/j.jchf.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/13/2024] [Accepted: 09/11/2024] [Indexed: 11/22/2024]
Abstract
The number of candidates on the waiting list for heart transplantation (HT) continues to far outweigh the number of available organs, and the donor heart nonuse rate in the United States remains significantly higher than that of other regions such as Europe. Although predicting outcomes in HT remains challenging, our overall understanding of the factors that play a role in post-HT outcomes continues to grow. We observe that many donor risk factors that are deemed "high-risk" do not necessarily always adversely affect post-HT outcomes, but are in fact nuanced and interact with other donor and recipient risk factors. The field of HT continues to evolve, with ongoing development of technologies for organ preservation during transport, expansion of the practice of donation after circulatory death, and proposed changes to organ allocation policy. As such, the field must continue to refine its processes for donor selection and risk prediction in HT.
Collapse
Affiliation(s)
- Rashmi Jain
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Evan P Kransdorf
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Jennifer Cowger
- Department of Cardiology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Valluvan Jeevanandam
- Department of Surgery, University of Chicago Medical Center, Chicago, Illinois, USA
| | - Jon A Kobashigawa
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
5
|
Karatasakis A, Kiamanesh O, Cheng RK, Kirkpatrick JN, Dudzinski DM. Echocardiographic Evaluation of the Post-Heart Transplant Patient. Curr Cardiol Rep 2025; 27:63. [PMID: 40014294 DOI: 10.1007/s11886-024-02169-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Significant practice variability exists with respect to the role and frequency of echocardiography after heart transplantation. We sought to illustrate key studies relating to the utility and diagnostic accuracy of echocardiography for the post-heart transplant patient. RECENT FINDINGS Several echocardiographic parameters correlate with acute heart transplant rejection, but there is enough heterogeneity between study results or in diagnostic accuracy, such that it precludes parameter use in isolation to rule out rejection. Newer techniques such as strain echocardiography may have better sensitivity. Similarly, resting and stress echocardiography can be combined with modern techniques such as myocardial contrast echocardiography to diagnose and prognosticate cardiac allograft vasculopathy, but studies have again demonstrated variable accuracy. Echocardiography remains an accessible tool in the evaluation and management of patients after heart transplantation. This modality can guide clinical judgment with real-time data and several fairly sensitive parameters for the detection of rejection, cardiac allograft vasculopathy, and other abnormalities. Often, auxiliary diagnostic modalities need to be combined to optimize diagnostic accuracy.
Collapse
Affiliation(s)
- Aris Karatasakis
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Omid Kiamanesh
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
| | - Richard K Cheng
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - James N Kirkpatrick
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Bioethics and Humanities, University of Washington, Seattle, WA, USA
| | - David M Dudzinski
- Section of Critical Care Cardiology and Echocardiography Laboratory, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.
| |
Collapse
|
6
|
Tsuji M, Patel JK, Kittleson MM, Chang DH, Kransdorf EP, Nikolova AP, Stern LK, Bhatnagar N, Kobashigawa JA. The outcome of restrictive cardiac allograft physiology in severe coronary allograft vasculopathy. J Heart Lung Transplant 2025; 44:57-63. [PMID: 39369967 DOI: 10.1016/j.healun.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Microvascular dysfunction after heart transplantation leads to restrictive cardiac allograft physiology (RCP), which is classified as severe coronary allograft vasculopathy (CAV); however, the prognosis of RCP remains unclear. Therefore, in this study, we aimed to elucidate the prognosis of RCP in comparison with that of severe angiographic CAV. METHODS We assessed 116 patients with severe CAV who underwent heart transplantation between 2004 and 2023. RCP was defined as symptomatic heart failure with restrictive hemodynamic values (mean right atrial pressure >12 mm Hg, pulmonary capillary wedge pressure >25 mm Hg, and cardiac index <2.0 liter/min/m2). The primary outcome was death or retransplantation. RESULTS Of the 116 patients with severe CAV, 42 had RCP (RCP-CAV group) and 74 had severe angiographic CAV without RCP (Angio-CAV group). A significantly shorter time from heart transplantation to diagnosis and lower subsequent percutaneous catheter intervention after diagnosis were seen in the RCP-CAV group than in the Angio-CAV group (both p < 0.001). Freedom from death or retransplantation at 5 years was significantly worse in the RCP-CAV group compared to the Angio-CAV group (18.4% vs 35.4%, p = 0.001). In the Cox proportional hazard model, RCP was independently associated with an increased risk of death or retransplantation (hazard ratio 2.08, 95% confidence intervals 1.26-3.44, p = 0.004). CONCLUSIONS The prognosis of patients with RCP was significantly worse than that of patients with severe angiographic CAV. The early detection of microvascular dysfunction and retransplantation listing may improve the prognosis of patients with RCP.
Collapse
Affiliation(s)
- Masaki Tsuji
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Jignesh K Patel
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - David H Chang
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Evan P Kransdorf
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Andriana P Nikolova
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Lily K Stern
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nayana Bhatnagar
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jon A Kobashigawa
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
7
|
Franco Acevedo A, Mack JJ, Valenzuela NM. The transcriptional repressor B cell lymphoma 6 regulates CXCR3 chemokine and human leukocyte antigen II expression in endothelial cells. Am J Transplant 2024; 24:2157-2173. [PMID: 39074669 DOI: 10.1016/j.ajt.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
Interferon gamma (IFN-γ) induces an endothelial proimmunogenic phenotype through the JAK/STAT1 pathway, which can shape the activation of alloreactive leukocytes in transplant rejection. In immune cells, the DNA-binding protein B cell lymphoma 6 (BCL6) controls the transcription of inflammatory genes. This study tested if BCL6 modulates IFN-γ-induced gene expression in endothelial cells. In vitro, BCL6 was IFN-γ-inducible in primary human endothelium, along with CXCR3 chemokines and human leukocyte antigen (HLA). BCL6, HLA II, and CXCL9 were also increased in human cardiac transplants during acute rejection. Knockdown of BCL6 augmented, whereas overexpression and BTB domain inhibitors (BCL6-BTBi) suppressed, HLA II and CXCR3 chemokine expression but not HLA I. Further, BCL6 had a greater effect on HLA-DR and DP but was less involved in regulating HLA-DQ expression. The effect correlated with BCL6 binding motifs in or near affected genes. The BCL6 DNA recognition sequence was highly similar to that of STAT1, and BTBi reduced STAT1's transcriptional activity in vitro. Our results show for the first time that BCL6 selectively controls IFN-γ-induced endothelial gene expression, advancing our understanding of the endogenous mechanisms regulating donor immunogenicity.
Collapse
Affiliation(s)
- Adriana Franco Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, USA
| | - Julia J Mack
- Department of Cardiology, University of California, Los Angeles, USA
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, USA.
| |
Collapse
|
8
|
Gamero MT, Brailovsky Y, Eisen HJ. Going the distance: Long-term follow-up of the randomized, prospective Scandinavian heart transplant everolimus de novo study with early calcineurin inhibitors avoidance (SCHEDULE) trial. J Heart Lung Transplant 2024; 43:1960-1962. [PMID: 39159701 DOI: 10.1016/j.healun.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024] Open
Affiliation(s)
- Maria T Gamero
- Jefferson Heart Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Yevgeniy Brailovsky
- Jefferson Heart Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Howard J Eisen
- Jefferson Heart Institute, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
9
|
Huang X, Yuzefpolskaya M, Colombo PC, Choe J, Shertel T, Jennings DL. The Impact of Statin Intensity on the Early Progression of Cardiac Allograft Vasculopathy. Clin Transplant 2024; 38:e70030. [PMID: 39558563 DOI: 10.1111/ctr.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/17/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Limited research has compared the relative risks and benefits different statins have after heart transplantation (HT). METHOD We hypothesize that higher statin intensity is associated with a smaller degree of allograft intimal thickening on intravascular ultrasound (IVUS) at 1-year post-HT. Allograft intima-media thickness (IMT) on the first annual IVUS was retrospectively compared in patients initiated on a low-intensity statin (pravastatin 20 mg daily) versus moderate-intensity statin (atorvastatin 20 mg daily) post-HT. RESULTS A total of 172 adult patients were included (2018-2022, n = 86 in each group). At 1 year, the maximal IMT was lower in the moderate-intensity statin group, but the difference did not reach statistical significance. The LDL levels at 1 year trended lower with moderate-intensity statin therapy, while the rates of adverse reactions were not statistically different. A multivariate analysis of the logistic regression model showed moderate statin intensity at 12 months was protective, while donor-specific antibodies developed within the first-year posttransplant were associated with IMT ≥ 0.5 mm on the first annual IVUS. CONCLUSION This study found that using moderate-intensity statin to prevent the early progression was as safe and possibly more effective than low-intensity statin therapy for the prevention of early cardiac allograft vasculopathy.
Collapse
Affiliation(s)
- Xinyi Huang
- Department of Pharmacy, New York Presbyterian Hospital, New York City, New York, USA
- Arnold & Marie Schwartz College of Pharmacy, Long Island University, Brooklyn, New York, USA
| | - Melana Yuzefpolskaya
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York City, New York, USA
| | - Paolo C Colombo
- Division of Cardiology, Department of Medicine, New York Presbyterian Hospital, Columbia University, New York City, New York, USA
| | - Jason Choe
- Department of Pharmacy, New York Presbyterian Hospital, New York City, New York, USA
| | - Tara Shertel
- Department of Pharmacy, New York Presbyterian Hospital, New York City, New York, USA
| | - Douglas L Jennings
- Department of Pharmacy, New York Presbyterian Hospital, New York City, New York, USA
- Arnold & Marie Schwartz College of Pharmacy, Long Island University, Brooklyn, New York, USA
| |
Collapse
|
10
|
Kim IC, Starling RC, Khush K, Passano E, Mirocha J, Bernhardt P, Azarbal B, Cheng R, Esmailian F, Mancini D, Patel JK, Sato T, Varnous S, Kobashigawa JA. Ten-year follow-up cohort of the everolimus versus azathioprine multinational prospective study focusing on intravascular ultrasound findings. J Heart Lung Transplant 2024; 43:1788-1794. [PMID: 39089607 DOI: 10.1016/j.healun.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 06/28/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Long-term clinical outcomes of early intravascular ultrasound (IVUS) findings in a prospective cohort of heart transplantation (HTx) patients have not been evaluated. METHODS This study included patients from 20 centers across Europe and North and South America among the original cohort of the RAD B253 study. Among these patients, 91 had paired IVUS images at baseline and 1-year post-transplant: everolimus 1.5 mg group (n = 25), everolimus 1.5 mg group (n = 33), and azathioprine 3.0 group (n = 33). The primary outcome was a composite of cardiovascular death, retransplantation, myocardial infarction (MI), coronary revascularization, and cardiac allograft vasculopathy (CAV) within a 10-year follow-up period. The secondary outcome was all-cause death, cardiovascular death, retransplantation, MI, coronary revascularization, and CAV. Donor disease was defined as baseline maximal intimal thickness (MIT) >0.66 mm, and rapid progression was defined as a change in MIT > 0.59 mm at 1 year. RESULTS Donor disease (46 patients) was associated with a higher incidence of the primary outcome (hazard ratio (HR) 4.444, 95% confidence interval [CI] 1.946-10.146, p < 0.001). Rapid progression (44 patients) was associated with a significantly higher incidence of the primary outcome (HR 2.942, 95% CI 1.383-6.260, p = 0.005). Higher-risk features on IVUS (positive both donor disease and rapid progression) were independently associated with poor clinical outcomes (HR 4.800, 95% CI 1.816-12.684, p = 0.002). CONCLUSIONS An increase in baseline MIT and a change in first-year MIT in IVUS post HTx was associated with poor outcomes up to 10 years. Early IVUS findings can be considered as surrogate endpoints for evaluating long-term outcomes in HTx clinical trials.
Collapse
Affiliation(s)
- In-Cheol Kim
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California; Division of Cardiology, Department of Internal Medicine, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Randall C Starling
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Kaufman Center for Heart Failure, Cleveland Clinic, Cleveland, Ohio
| | - Kiran Khush
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Elizabeth Passano
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - James Mirocha
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Peter Bernhardt
- Department of Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - Babak Azarbal
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Richard Cheng
- Cardiology Division, University of Washington, Seattle, Washington
| | - Fardad Esmailian
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Donna Mancini
- Cardiology Department, Cardiovascular Institute, Mount Sinai Hospital, New York, New York
| | - Jignesh K Patel
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Takuma Sato
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shaida Varnous
- Department of Thoracic and Cardiovascular Surgery, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Jon A Kobashigawa
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
11
|
Peled Y, Ducharme A, Kittleson M, Bansal N, Stehlik J, Amdani S, Saeed D, Cheng R, Clarke B, Dobbels F, Farr M, Lindenfeld J, Nikolaidis L, Patel J, Acharya D, Albert D, Aslam S, Bertolotti A, Chan M, Chih S, Colvin M, Crespo-Leiro M, D'Alessandro D, Daly K, Diez-Lopez C, Dipchand A, Ensminger S, Everitt M, Fardman A, Farrero M, Feldman D, Gjelaj C, Goodwin M, Harrison K, Hsich E, Joyce E, Kato T, Kim D, Luong ML, Lyster H, Masetti M, Matos LN, Nilsson J, Noly PE, Rao V, Rolid K, Schlendorf K, Schweiger M, Spinner J, Townsend M, Tremblay-Gravel M, Urschel S, Vachiery JL, Velleca A, Waldman G, Walsh J. International Society for Heart and Lung Transplantation Guidelines for the Evaluation and Care of Cardiac Transplant Candidates-2024. J Heart Lung Transplant 2024; 43:1529-1628.e54. [PMID: 39115488 DOI: 10.1016/j.healun.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 08/18/2024] Open
Abstract
The "International Society for Heart and Lung Transplantation Guidelines for the Evaluation and Care of Cardiac Transplant Candidates-2024" updates and replaces the "Listing Criteria for Heart Transplantation: International Society for Heart and Lung Transplantation Guidelines for the Care of Cardiac Transplant Candidates-2006" and the "2016 International Society for Heart Lung Transplantation Listing Criteria for Heart Transplantation: A 10-year Update." The document aims to provide tools to help integrate the numerous variables involved in evaluating patients for transplantation, emphasizing updating the collaborative treatment while waiting for a transplant. There have been significant practice-changing developments in the care of heart transplant recipients since the publication of the International Society for Heart and Lung Transplantation (ISHLT) guidelines in 2006 and the 10-year update in 2016. The changes pertain to 3 aspects of heart transplantation: (1) patient selection criteria, (2) care of selected patient populations, and (3) durable mechanical support. To address these issues, 3 task forces were assembled. Each task force was cochaired by a pediatric heart transplant physician with the specific mandate to highlight issues unique to the pediatric heart transplant population and ensure their adequate representation. This guideline was harmonized with other ISHLT guidelines published through November 2023. The 2024 ISHLT guidelines for the evaluation and care of cardiac transplant candidates provide recommendations based on contemporary scientific evidence and patient management flow diagrams. The American College of Cardiology and American Heart Association modular knowledge chunk format has been implemented, allowing guideline information to be grouped into discrete packages (or modules) of information on a disease-specific topic or management issue. Aiming to improve the quality of care for heart transplant candidates, the recommendations present an evidence-based approach.
Collapse
Affiliation(s)
- Yael Peled
- Leviev Heart & Vascular Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Anique Ducharme
- Deparment of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada.
| | - Michelle Kittleson
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Neha Bansal
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Josef Stehlik
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Shahnawaz Amdani
- Department of Pediatric Cardiology, Cleveland Clinic Children's, Cleveland, Ohio, USA
| | - Diyar Saeed
- Heart Center Niederrhein, Helios Hospital Krefeld, Krefeld, Germany
| | - Richard Cheng
- Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Brian Clarke
- Division of Cardiology, University of British Columbia, St Paul's Hospital, Vancouver, British Columbia, Canada
| | - Fabienne Dobbels
- Academic Centre for Nursing and Midwifery, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Maryjane Farr
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX; Parkland Health System, Dallas, TX, USA
| | - JoAnn Lindenfeld
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | | | - Jignesh Patel
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Deepak Acharya
- Division of Cardiovascular Diseases, University of Arizona Sarver Heart Center, Tucson, Arizona, USA
| | - Dimpna Albert
- Department of Paediatric Cardiology, Paediatric Heart Failure and Cardiac Transplant, Heart Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Saima Aslam
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alejandro Bertolotti
- Heart and Lung Transplant Service, Favaloro Foundation University Hospital, Buenos Aires, Argentina
| | - Michael Chan
- University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Sharon Chih
- Heart Failure and Transplantation, Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Monica Colvin
- Department of Cardiology, University of Michigan, Ann Arbor, MI; Scientific Registry of Transplant Recipients, Hennepin Healthcare Research Institute, Minneapolis, MN, USA
| | - Maria Crespo-Leiro
- Cardiology Department Complexo Hospitalario Universitario A Coruna (CHUAC), CIBERCV, INIBIC, UDC, La Coruna, Spain
| | - David D'Alessandro
- Massachusetts General Hospital, Boston; Harvard School of Medicine, Boston, MA, USA
| | - Kevin Daly
- Boston Children's Hospital & Harvard Medical School, Boston, MA, USA
| | - Carles Diez-Lopez
- Advanced Heart Failure and Heart Transplant Unit, Department of Cardiology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Anne Dipchand
- Division of Cardiology, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | - Melanie Everitt
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Alexander Fardman
- Leviev Heart & Vascular Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Marta Farrero
- Department of Cardiology, Hospital Clínic, Barcelona, Spain
| | - David Feldman
- Newark Beth Israel Hospital & Rutgers University, Newark, NJ, USA
| | - Christiana Gjelaj
- Department of Cardiovascular and Thoracic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Matthew Goodwin
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, UT, USA
| | - Kimberly Harrison
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eileen Hsich
- Cleveland Clinic Foundation, Division of Cardiovascular Medicine, Cleveland, OH, USA
| | - Emer Joyce
- Department of Cardiology, Mater University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| | - Tomoko Kato
- Department of Cardiology, International University of Health and Welfare School of Medicine, Narita, Chiba, Japan
| | - Daniel Kim
- University of Alberta & Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Me-Linh Luong
- Division of Infectious Disease, Department of Medicine, University of Montreal Hospital Center, Montreal, Quebec, Canada
| | - Haifa Lyster
- Department of Heart and Lung Transplantation, The Royal Brompton and Harefield NHS Foundation Trust, Harefield Hospital, Harefield, Middlesex, UK
| | - Marco Masetti
- Heart Failure and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Johan Nilsson
- Department of Cardiothoracic and Vascular Surgery, Skane University Hospital, Lund, Sweden
| | | | - Vivek Rao
- Division of Cardiac Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Katrine Rolid
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kelly Schlendorf
- Division of Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Joseph Spinner
- Section of Pediatric Cardiology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Madeleine Townsend
- Division of Pediatric Cardiology, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Maxime Tremblay-Gravel
- Deparment of Medicine, Montreal Heart Institute, Université?de Montréal, Montreal, Quebec, Canada
| | - Simon Urschel
- Stollery Children's Hospital, University of Alberta, Edmonton, Alberta, Canada
| | - Jean-Luc Vachiery
- Department of Cardiology, Cliniques Universitaires de Bruxelles, Hôpital Académique Erasme, Bruxelles, Belgium
| | - Angela Velleca
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Georgina Waldman
- Department of Pharmacy, Massachusetts General Hospital, Boston, MA, USA
| | - James Walsh
- Allied Health Research Collaborative, The Prince Charles Hospital, Brisbane; Heart Lung Institute, The Prince Charles Hospital, Brisbane, Australia
| |
Collapse
|
12
|
Nikolova AP, Kobashigawa JA. The EVOLVeD Role of Cholesterol-Lowering Therapies in Cardiac Allograft Vasculopathy. JACC. HEART FAILURE 2024; 12:1689-1691. [PMID: 39023491 DOI: 10.1016/j.jchf.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 07/20/2024]
Affiliation(s)
- Andriana P Nikolova
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| | - Jon A Kobashigawa
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
13
|
Harris E, Prasad N, Skoll D, Kumar SS, Fried J, Topkara V, Raikhelkar JK, DeFilippis EM, Latif F, Yuzefpolskaya M, Colombo PC, Uriel N, Takeda K, Sayer GT, Clerkin KJ. CAV Trajectories Among Patients With No or Mild CAV at 10 Years Posttransplant. Clin Transplant 2024; 38:e70009. [PMID: 39436145 DOI: 10.1111/ctr.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/29/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024]
Abstract
Cardiac allograft vasculopathy (CAV) is a major cause of morbidity and mortality following heart transplantation (HT). Prior studies identified distinct CAV trajectories in the early post-HT period with unique predictors, but the evolution of CAV in later periods is not well-described. This study assessed the prevalence of late CAV progression and associated risk factors in HT recipients with ISHLT CAV 0/1 at 10 years post-HT. Consecutive adult patients who underwent HT from January 2000 to December 2008 were evaluated and grouped by CAV trajectories into progressors (developed ISHLT CAV 2/3) or nonprogressors (remained ISHLT CAV 0/1). A total of 130 patients were included with a median age at angiography of 61.7 years and a median follow-up time of 4.8 years. 8.5% progressed to CAV 2/3, while the remaining 91.5% were nonprogressors. Progression was not associated with death or retransplantation (27.3% [progressor] vs. 21.0% [nonprogressor], p = 0.70). These data may inform shared decision-making about late CAV screening.
Collapse
Affiliation(s)
- Erin Harris
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Nikil Prasad
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Devin Skoll
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Sambhavi Sneha Kumar
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Justin Fried
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Veli Topkara
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Jayant K Raikhelkar
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Ersilia M DeFilippis
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Farhana Latif
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Melana Yuzefpolskaya
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Paolo C Colombo
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Nir Uriel
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Koji Takeda
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, NewYork-Presbyterian/Columbia University Medical Center, New York, New York, USA
| | - Gabriel T Sayer
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| | - Kevin J Clerkin
- Milstein Division of Cardiology, Department of Medicine, NewYork-Presbyterian/Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
14
|
Naimimohasses S, Keshavjee S, Wang B, Brudno M, Sidhu A, Bhat M. Proceedings of the 2024 Transplant AI Symposium. FRONTIERS IN TRANSPLANTATION 2024; 3:1399324. [PMID: 39319335 PMCID: PMC11421390 DOI: 10.3389/frtra.2024.1399324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024]
Abstract
With recent advancements in deep learning (DL) techniques, the use of artificial intelligence (AI) has become increasingly prevalent in all fields. Currently valued at 9.01 billion USD, it is a rapidly growing market, projected to increase by 40% per annum. There has been great interest in how AI could transform the practice of medicine, with the potential to improve all healthcare spheres from workflow management, accessibility, and cost efficiency to enhanced diagnostics with improved prognostic accuracy, allowing the practice of precision medicine. The applicability of AI is particularly promising for transplant medicine, in which it can help navigate the complex interplay of a myriad of variables and improve patient care. However, caution must be exercised when developing DL models, ensuring they are trained with large, reliable, and diverse datasets to minimize bias and increase generalizability. There must be transparency in the methodology and extensive validation of the model, including randomized controlled trials to demonstrate performance and cultivate trust among physicians and patients. Furthermore, there is a need to regulate this rapidly evolving field, with updated policies for the governance of AI-based technologies. Taking this in consideration, we summarize the latest transplant AI developments from the Ajmera Transplant Center's inaugural symposium.
Collapse
Affiliation(s)
- Sara Naimimohasses
- Division of Gastroenterology, Toronto General Hospital, Toronto, ON, Canada
- Ajmera Transplant Center, University Health Network, Toronto, ON, Canada
| | - Shaf Keshavjee
- Department of Innovation, University Health Network, Toronto, ON, Canada
| | - Bo Wang
- Department of Laboratory Medicine and Pathobiology, The Temerty Centre for AI Research and Education in Medicine, Toronto, ON, Canada
| | - Mike Brudno
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Aman Sidhu
- Division of Gastroenterology, Toronto General Hospital, Toronto, ON, Canada
- Ajmera Transplant Center, University Health Network, Toronto, ON, Canada
| | - Mamatha Bhat
- Division of Gastroenterology, Toronto General Hospital, Toronto, ON, Canada
- Ajmera Transplant Center, University Health Network, Toronto, ON, Canada
| |
Collapse
|
15
|
Farcas AO, Stoica MC, Maier IM, Maier AC, Sin AI. Heart Transplant Rejection: From the Endomyocardial Biopsy to Gene Expression Profiling. Biomedicines 2024; 12:1926. [PMID: 39200392 PMCID: PMC11351478 DOI: 10.3390/biomedicines12081926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/02/2024] Open
Abstract
Heart transplant prolongs life for patients with end-stage heart failure but rejection remains a complication that reduces long-term survival. The aim is to provide a comprehensive overview of the current status in HT rejection. EMB is an invasive diagnostic tool, consisting in the sampling of a fragment of myocardial tissue from the right ventricular septum using fluoroscopic guidance. This tissue can later be subjected to histopathological, immunohistochemical or molecular analysis, providing valuable information for cardiac allograft rejection, but this procedure is not without complications. To increase the accuracy of the rejection diagnosis, EMB requires a systematic evaluation of endocardium, myocardium, interstitium and intramural vessels. There are three types of rejection: hyperacute, acute or chronic, diagnosed by the histopathological evaluation of EMB as well as by new diagnostic methods such as DSA, ddcfDNA and gene expression profiling, the last having a high negative predictive value. More than 50 years after the introduction of EMB in medical practice, it still remains the "gold standard" in monitoring rejection in HT recipients but other new, less invasive diagnostic methods reduce the number of EMBs required.
Collapse
Affiliation(s)
- Anca Otilia Farcas
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Department of Cell Biology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Mihai Ciprian Stoica
- Department of Nephrology/Internal Medicine, Mures County Clinical Hospital, 540103 Targu Mures, Romania
- Department of Internal Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania
| | | | - Adrian Cornel Maier
- Emergency Military Hospital, 800150 Galati, Romania;
- Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800008 Galati, Romania
| | - Anca Ileana Sin
- Department of Cell Biology, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Department of Pathology, Clinical County Emergency Hospital, 540136 Targu Mures, Romania
| |
Collapse
|
16
|
Severo Sánchez A, González Martín J, de Juan Bagudá J, Morán Fernández L, Muñoz Guijosa C, Arribas Ynsaurriaga F, Delgado JF, García-Cosío Carmena MD. Sex and Gender-related Disparities in Clinical Characteristics and Outcomes in Heart Transplantation. Curr Heart Fail Rep 2024; 21:367-378. [PMID: 38861129 DOI: 10.1007/s11897-024-00670-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
PURPOSE OF REVIEW Limited research has been conducted on sex disparities in heart transplant (HT). The aim of this review is to analyse the available evidence on the influence of sex and gender-related determinants in the entire HT process, as well as to identify areas for further investigation. RECENT FINDINGS Although women make up half of the population affected by heart failure and related mortality, they account for less than a third of HT recipients. Reasons for this inequality include differences in disease course, psychosocial factors, concerns about allosensitisation, and selection or referral bias in female patients. Women are more often listed for HT due to non-ischaemic cardiomyopathy and have a lower burden of cardiovascular risk factors. Although long-term prognosis appears to be similar for both sexes, there are significant disparities in post-HT morbidity and causes of mortality (noting a higher incidence of rejection in women and of malignancy and cardiac allograft vasculopathy in men). Additional research is required to gain a better understanding of the reasons behind gender disparities in eligibility and outcomes following HT. This would enable the fair allocation of resources and enhance patient care.
Collapse
Affiliation(s)
- Andrea Severo Sánchez
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Javier González Martín
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Javier de Juan Bagudá
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Laura Morán Fernández
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
| | - Christian Muñoz Guijosa
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
- Cardiac Surgery Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Arribas Ynsaurriaga
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Juan Francisco Delgado
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - María Dolores García-Cosío Carmena
- Cardiology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.
- Centro Nacional de Investigaciones Biomédicas en Red de Enfermedades CardioVasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
17
|
Halloran PF, Madill-Thomsen K, Aliabadi-Zuckermann AZ, Cadeiras M, Crespo-Leiro MG, Depasquale EC, Deng M, Gökler J, Hall S, Jamil A, Kim DH, Kobashigawa J, Macdonald P, Melenovsky V, Patel J, Potena L, Shah K, Stehlik J, Zuckermann A. Redefining the molecular rejection states in 3230 heart transplant biopsies: Relationships to parenchymal injury and graft survival. Am J Transplant 2024; 24:1414-1426. [PMID: 38527588 DOI: 10.1016/j.ajt.2024.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
The first-generation Molecular Microscope (MMDx) system for heart transplant endomyocardial biopsies used expression of rejection-associated transcripts (RATs) to diagnose not only T cell-mediated rejection (TCMR) and antibody-mediated rejection (ABMR) but also acute injury. However, the ideal system should detect rejection without being influenced by injury, to permit analysis of the relationship between rejection and parenchymal injury. To achieve this, we developed a new rejection classification in an expanded cohort of 3230 biopsies: 1641 from INTERHEART (ClinicalTrials.gov NCT02670408), plus 1589 service biopsies added to improve the power of the machine learning algorithms. The new system used 6 rejection classifiers instead of RATs and generated 7 rejection archetypes: No rejection, 48%; Minor, 24%; TCMR1, 2.3%; TCMR2, 2.7%; TCMR/mixed, 2.7%; early-stage ABMR, 3.9%; and fully developed ABMR, 16%. Using rejection classifiers eliminated cross-reactions with acute injury, permitting separate assessment of rejection and injury. TCMR was associated with severe-recent injury and late atrophy-fibrosis and rarely had normal parenchyma. ABMR was better tolerated, seldom producing severe injury, but in later biopsies was often associated with atrophy-fibrosis, indicating long-term risk. Graft survival and left ventricular ejection fraction were reduced not only in hearts with TCMR but also in hearts with severe-recent injury and atrophy-fibrosis, even without rejection.
Collapse
Affiliation(s)
- Philip F Halloran
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| | | | | | - Martin Cadeiras
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
| | - Marisa G Crespo-Leiro
- Advanced Heart Failure and Heart Transplant Unit, Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | | | - Mario Deng
- Ronald Reagan UCLA Medical Center, Los Angeles, California, USA
| | - Johannes Gökler
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Aayla Jamil
- Baylor Scott & White Health, Dallas, Texas, USA
| | - Daniel H Kim
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Jon Kobashigawa
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter Macdonald
- The Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Vojtech Melenovsky
- Department of Cardiology, Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Jignesh Patel
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Luciano Potena
- Heart Failure and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Keyur Shah
- Department of Cardiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Josef Stehlik
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Andreas Zuckermann
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Alessandrini A, Rosales IA. Down to the coronary arteries: When spatial multiomics open up a narrow path. Am J Transplant 2024; 24:1102-1104. [PMID: 38508320 DOI: 10.1016/j.ajt.2024.02.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Affiliation(s)
- Alessandro Alessandrini
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ivy A Rosales
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Pathology, Immunopathology Research Laboratory, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
19
|
Wall C, Weir-McCall J, Tweed K, Hoole SP, Gopalan D, Huang Y, Corovic A, Peverelli M, Dey D, Bennett MR, Rudd JHF, Kydd A, Bhagra S, Tarkin JM. Computed tomography pericoronary adipose tissue density predicts coronary allograft vasculopathy and adverse clinical outcomes after cardiac transplantation. Eur Heart J Cardiovasc Imaging 2024; 25:1018-1027. [PMID: 38493483 PMCID: PMC11210971 DOI: 10.1093/ehjci/jeae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024] Open
Abstract
AIMS To assess pericoronary adipose tissue (PCAT) density on coronary computed tomography angiography (CCTA) as a marker of inflammatory disease activity in coronary allograft vasculopathy (CAV). METHODS AND RESULTS PCAT density, lesion volumes, and total vessel volume-to-myocardial mass ratio (V/M) were retrospectively measured in 126 CCTAs from 94 heart transplant patients [mean age 49 (SD 14.5) years, 40% female] who underwent imaging between 2010 and 2021; age- and sex-matched controls; and patients with atherosclerosis. PCAT density was higher in transplant patients with CAV [n = 40; -73.0 HU (SD 9.3)] than without CAV [n = 86; -77.9 HU (SD 8.2)], and controls [n = 12; -86.2 HU (SD 5.4)], P < 0.01 for both. Unlike patients with atherosclerotic coronary artery disease (n = 32), CAV lesions were predominantly non-calcified and comprised of mostly fibrous or fibrofatty tissue. V/M was lower in patients with CAV than without [32.4 mm3/g (SD 9.7) vs. 41.4 mm3/g (SD 12.3), P < 0.0001]. PCAT density and V/M improved the ability to predict CAV from area under the receiver operating characteristic curve (AUC) 0.75-0.85 when added to donor age and donor hypertension status (P < 0.0001). PCAT density above -66 HU was associated with a greater incidence of all-cause mortality {odds ratio [OR] 18.0 [95% confidence interval (CI) 3.25-99.6], P < 0.01} and the composite endpoint of death, CAV progression, acute rejection, and coronary revascularization [OR 7.47 (95% CI 1.8-31.6), P = 0.01] over 5.3 (SD 2.1) years. CONCLUSION Heart transplant patients with CAV have higher PCAT density and lower V/M than those without. Increased PCAT density is associated with adverse clinical outcomes. These CCTA metrics could be useful for the diagnosis and monitoring of CAV severity.
Collapse
Affiliation(s)
- Christopher Wall
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Jonathan Weir-McCall
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Radiology, Royal Papworth Hospital, Cambridge, UK
| | - Katharine Tweed
- Department of Radiology, Royal Papworth Hospital, Cambridge, UK
| | - Stephen P Hoole
- Department of Cardiology, Royal Papworth Hospital, Cambridge, UK
| | - Deepa Gopalan
- Department of Radiology, Cambridge University Hospitals NHS Trust, Cambridge, UK
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - Yuan Huang
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Andrej Corovic
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Marta Peverelli
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Damini Dey
- Departments of Biomedical Sciences and Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
| | - Martin R Bennett
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - James H F Rudd
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Anna Kydd
- Transplant Unit, Royal Papworth Hospital, Cambridge, UK
| | - Sai Bhagra
- Transplant Unit, Royal Papworth Hospital, Cambridge, UK
| | - Jason M Tarkin
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| |
Collapse
|
20
|
Demir Z, Raynaud M, Aubert O, Debray D, Sebagh M, Duong Van Huyen JP, Del Bello A, Jolivet NC, Paradis V, Durand F, Muratot S, Lozach C, Chardot C, Francoz C, Kamar N, Sarnacki S, Coilly A, Samuel D, Vibert E, Féray C, Lefaucheur C, Loupy A. Identification of liver transplant biopsy phenotypes associated with distinct liver biological markers and allograft survival. Am J Transplant 2024; 24:954-966. [PMID: 38097016 DOI: 10.1016/j.ajt.2023.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/07/2023] [Accepted: 12/06/2023] [Indexed: 01/01/2024]
Abstract
The intricate association between histologic lesions and circulating antihuman leucocyte antigen donor-specific antibodies (DSA) in liver transplantation (LT) requires further clarification. We conducted a probabilistic, unsupervised approach in a comprehensively well-annotated LT cohort to identify clinically relevant archetypes. We evaluated 490 pairs of LT biopsies with DSA testing from 325 recipients transplanted between 2010 and 2020 across 3 French centers and an external cohort of 202 biopsies from 128 recipients. Unsupervised archetypal analysis integrated all clinico-immuno-histologic parameters of each biopsy to identify biopsy archetypes. The median time after LT was 1.17 (interquartile range, 0.38-2.38) years. We identified 7 archetypes distinguished by clinico-immuno-histologic parameters: archetype #1: severe T cell-mediated rejection (15.9%); #2: chronic rejection with ductopenia (1.8%); #3: architectural and microvascular damages (3.5%); #4: (sub)normal (55.9%); #5: mild T cell-mediated rejection (4.9%); #6: acute antibody-mediated rejection (6.5%); and #7: chronic rejection with DSA (11.4%). Cell infiltrates vary in the archetype. These archetypes were associated with distinct liver biological markers and allograft outcomes. These findings remained consistent when stratified using the patient's age or indications for LT, with good performance in the external cohort (mean highest probability assignment = 0.58, standard deviation ± 0.17). In conclusion, we have identified clinically meaningful archetypes, providing valuable insights into the intricate DSA-histology association, which may help standardize liver allograft pathology classification.
Collapse
Affiliation(s)
- Zeynep Demir
- Paris Translational Research Center for Organ Transplantation, Université de Paris Cité, INSERM, PARCC, Paris, France
| | - Marc Raynaud
- Paris Translational Research Center for Organ Transplantation, Université de Paris Cité, INSERM, PARCC, Paris, France
| | - Olivier Aubert
- Paris Translational Research Center for Organ Transplantation, Université de Paris Cité, INSERM, PARCC, Paris, France; Kidney Transplantation Department, Necker enfants malades Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Dominique Debray
- Pediatric Hepatology and Liver Transplantation Unit, Necker enfants malades Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Mylène Sebagh
- Pathology Department Paul-Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Villejuif, France
| | - Jean-Paul Duong Van Huyen
- Pathology Department, Necker enfants malades Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Arnaud Del Bello
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France
| | - Nicolas Congy Jolivet
- Department of Immunology, Hôpital de Rangueil, CHU de Toulouse, Molecular Immunogenetics Laboratory, EA 3034, IFR150 (INSERM), Toulouse, France
| | - Valérie Paradis
- Pathology Department, Beaujon Hospital, Assistance Publique - Hôpitaux de Paris, Clichy, France
| | - François Durand
- Hepatology Department, Beaujon Hospital, Assistance Publique - Hôpitaux de Paris, Clichy, France
| | - Sophie Muratot
- Paris Translational Research Center for Organ Transplantation, Université de Paris Cité, INSERM, PARCC, Paris, France
| | - Cécile Lozach
- Department of Pediatric Radiology, Necker enfants malades Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Christophe Chardot
- Department of Pediatric Surgery, Necker enfants malades Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Claire Francoz
- Hepatology Department, Beaujon Hospital, Assistance Publique - Hôpitaux de Paris, Clichy, France
| | - Nassim Kamar
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France
| | - Sabine Sarnacki
- Department of Pediatric Surgery, Necker enfants malades Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Audrey Coilly
- Hepatobiliary Center, Paul-Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Inserm Paris-Saclay Research Unit 1193, Paris-Saclay University, Villejuif, France
| | - Didier Samuel
- Hepatobiliary Center, Paul-Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Inserm Paris-Saclay Research Unit 1193, Paris-Saclay University, Villejuif, France
| | - Eric Vibert
- Hepatobiliary Center, Paul-Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Inserm Paris-Saclay Research Unit 1193, Paris-Saclay University, Villejuif, France
| | - Cyrille Féray
- Hepatobiliary Center, Paul-Brousse Hospital, Assistance Publique - Hôpitaux de Paris, Inserm Paris-Saclay Research Unit 1193, Paris-Saclay University, Villejuif, France
| | - Carmen Lefaucheur
- Paris Translational Research Center for Organ Transplantation, Université de Paris Cité, INSERM, PARCC, Paris, France; Department of Nephrology and Kidney Transplantation, Saint-Louis Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Alexandre Loupy
- Paris Translational Research Center for Organ Transplantation, Université de Paris Cité, INSERM, PARCC, Paris, France; Kidney Transplantation Department, Necker enfants malades Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France.
| |
Collapse
|
21
|
Abadie B, Albert C, Bhat P, Harb S, Jacob M, Starling RC, Tang WHW, Jaber WA. Frequency of screening for cardiac allograft vasculopathy: warranty period of initial low risk positron emission tomography. Eur Heart J Cardiovasc Imaging 2024; 25:814-820. [PMID: 38214683 DOI: 10.1093/ehjci/jeae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/13/2024] Open
Abstract
AIMS The short-term risk of moderate-severe cardiac allograft vasculopathy (CAV) after a low-risk positron emission tomography/computed tomography (PET/CT) is unknown, and therefore, there is no guidance on how frequently to perform screening. The aim of this study was to assess the rate of progression to moderate-severe CAV as part of an annual screening programme. METHODS AND RESULTS Patients with no history of CAV 2/3 and a low-risk result on initial screening PET/CT (CAV 0/1) were enrolled in the study. The primary outcome was the progression to CAV 2/3 as part of an annual screening programme (within 6-18 months of initial scan). PET CAV results were graded according to a published and externally validated diagnostic criterion for CAV. Over the study period, 231 patients underwent an initial PET/CT and had a subsequent evaluation for CAV. In this cohort, 4.3% of patients progressed to CAV 2/3 at a median of 374 days (interquartile range 363-433). Initial PET CAV grade was the most significant patient characteristic associated with the progression of CAV, with 17% of patients with PET CAV 1 progressing to CAV 2/3 compared with 1.6% with PET CAV 0 (odds ratio 12.4, 95% confidence interval 3.06-50.3). CONCLUSION The rate of progression to moderate-severe CAV at 1 year after the lowest-risk PET/CT is low, but approximately 1/6 patients with PET CAV 1 progress to CAV 2/3. Annual screening with PET/CT for select patients with PET CAV 0 may not be warranted. The optimal screening interval awaits confirmation of our findings in multi-centre registries.
Collapse
Affiliation(s)
- Bryan Abadie
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| | - Chonyang Albert
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| | - Pavan Bhat
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| | - Serge Harb
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| | - Miriam Jacob
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| | - Randall C Starling
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| | - W H Wilson Tang
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| | - Wael A Jaber
- Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Avenue, J1-5 Main Campus, Cleveland, OH 44195, USA
| |
Collapse
|
22
|
Amancherla K, Feurer ID, Rega SA, Cluckey A, Salih M, Davis J, Pedrotty D, Ooi H, Rali AS, Siddiqi HK, Menachem J, Brinkley DM, Punnoose L, Sacks SB, Zalawadiya SK, Wigger M, Balsara K, Trahanas J, McMaster WG, Hoffman J, Pasrija C, Lindenfeld J, Shah AS, Schlendorf KH. Early Assessment of Cardiac Allograft Vasculopathy Risk Among Recipients of Hepatitis C Virus-infected Donors in the Current Era. J Card Fail 2024; 30:694-700. [PMID: 37907147 PMCID: PMC11056484 DOI: 10.1016/j.cardfail.2023.09.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Transplantation of hearts from hepatitis C virus (HCV)-positive donors has increased substantially in recent years following development of highly effective direct-acting antiviral therapies for treatment and cure of HCV. Although historical data from the pre-direct-acting antiviral era demonstrated an association between HCV-positive donors and accelerated cardiac allograft vasculopathy (CAV) in recipients, the relationship between the use of HCV nucleic acid test-positive (NAT+) donors and the development of CAV in the direct-acting antiviral era remains unclear. METHODS AND RESULTS We performed a retrospective, single-center observational study comparing coronary angiographic CAV outcomes during the first year after transplant in 84 heart transplant recipients of HCV NAT+ donors and 231 recipients of HCV NAT- donors. Additionally, in a subsample of 149 patients (including 55 in the NAT+ cohort and 94 in the NAT- cohort) who had serial adjunctive intravascular ultrasound examination performed, we compared development of rapidly progressive CAV, defined as an increase in maximal intimal thickening of ≥0.5 mm in matched vessel segments during the first year post-transplant. In an unadjusted analysis, recipients of HCV NAT+ hearts had reduced survival free of CAV ≥1 over the first year after heart transplant compared with recipients of HCV NAT- hearts. After adjustment for known CAV risk factors, however, there was no significant difference between cohorts in the likelihood of the primary outcome, nor was there a difference in development of rapidly progressive CAV. CONCLUSIONS These findings support larger, longer-term follow-up studies to better elucidate CAV outcomes in recipients of HCV NAT+ hearts and to inform post-transplant management strategies.
Collapse
Affiliation(s)
- Kaushik Amancherla
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Irene D Feurer
- Departments of Surgery and Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Scott A Rega
- Vanderbilt Transplant Center, Nashville, Tennessee
| | - Andrew Cluckey
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mohamed Salih
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan Davis
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dawn Pedrotty
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Henry Ooi
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Aniket S Rali
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hasan K Siddiqi
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jonathan Menachem
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas M Brinkley
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynn Punnoose
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Suzanne B Sacks
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sandip K Zalawadiya
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mark Wigger
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Keki Balsara
- Department of Cardiac Surgery, Medstar Washington Hospital Center, Washington, DC
| | - John Trahanas
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - William G McMaster
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jordan Hoffman
- Division of Cardiothoracic Surgery, University of Colorado, Aurora, Colorado
| | - Chetan Pasrija
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joann Lindenfeld
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ashish S Shah
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kelly H Schlendorf
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
23
|
Birs AS, Bui QM, Gernhofer Y, Duran A, Keyt L, Paternostro K, Ding J, Adler E, Ang L, Urey MA, Kearns MJ, Wettersten N, Pretorius V. Cardiac allograft vasculopathy outcomes among donation after circulatory death heart transplant recipients. JHLT OPEN 2024; 4:100065. [PMID: 40144269 PMCID: PMC11935384 DOI: 10.1016/j.jhlto.2024.100065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background Cardiac allograft vasculopathy (CAV) accounts for significant long-term morbidity and mortality in heart transplant recipients; limited data exist for donation after circulatory death (DCD). Intravascular ultrasound (IVUS) assessment is a gold standard for early diagnosis of CAV and has strong prognostic power. Methods We evaluated all consecutive circulatory and brain death heart transplant recipients from January 2020 to March 2022. Patients were followed for need for percutaneous coronary intervention (PCI), development of severe allograft vasculopathy, or death. Among 143 heart transplant recipients, 39 received circulatory death and 104 received brain death hearts. Results Baseline characteristics were similar between groups: median age (56.3 vs 53.7 years, p = 0.290), female sex (15% vs 26%, p = 0.265), and sirolimus use (69% vs 53%, p = 0.116). At 1 year, there were no significant differences in maximal intimal thickness (0.49 vs 0.46 mm, p = 0.861) or Stanford classification. During a median follow-up of 793 days [interquartile ranges 618, 1003], there was no difference in the unadjusted or adjusted primary composite outcome of death, PCI, or International Society of Heart and Lung Transplantation cardiac allograft vasculopathy maximal intimal thickness ≥0.6 mm (unadjusted hazard ratio (HR) 0.42, 95% confidence interval (CI): 0.05, 3.48, p = 0.42), event rate 9.6% vs 2.6%, p = 0.29, nor was there a difference in death, PCI or severe IVUS disease (HR 1.44, 95% CI 0.81, 2.56, p = 0.21). Conclusion In DCD heart transplant recipients, circulatory death donors did not have a significantly higher risk for coronary allograft vasculopathy by IVUS or related complications at 1 year following transplantation.
Collapse
Affiliation(s)
- Antoinette S. Birs
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Quan M. Bui
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Yan Gernhofer
- University of the Incarnate Word School of Osteopathic Medicine, San Antonio, Texas
| | - Antonio Duran
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Lucas Keyt
- University of California San Diego School of Medicine, La Jolla, California
| | - Kevin Paternostro
- University of California San Diego School of Medicine, La Jolla, California
| | - Jeffrey Ding
- University of California San Diego School of Medicine, La Jolla, California
| | - Eric Adler
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Lawrence Ang
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Marcus A. Urey
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Mark J. Kearns
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| | - Nicholas Wettersten
- Division of Cardiovascular Medicine, San Diego Veterans Affairs Medical Center, San Diego, California
| | - Victor Pretorius
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| |
Collapse
|
24
|
Shah P, Agbor-Enoh S, Lee S, Andargie TE, Sinha SS, Kong H, Henry L, Park W, McNair E, Tchoukina I, Shah KB, Najjar SS, Hsu S, Rodrigo ME, Jang MK, Marboe C, Berry GJ, Valantine HA. Racial Differences in Donor-Derived Cell-Free DNA and Mitochondrial DNA After Heart Transplantation, on Behalf of the GRAfT Investigators. Circ Heart Fail 2024; 17:e011160. [PMID: 38375637 PMCID: PMC11021168 DOI: 10.1161/circheartfailure.123.011160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/07/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Black heart transplant patients are at higher risk of acute rejection (AR) and death than White patients. We hypothesized that this risk may be associated with higher levels of donor-derived cell-free DNA (dd-cfDNA) and cell-free mitochondrial DNA. METHODS The Genomic Research Alliance for Transplantation is a multicenter, prospective, longitudinal cohort study. Sequencing was used to quantitate dd-cfDNA and polymerase chain reaction to quantitate cell-free mitochondrial DNA in plasma. AR was defined as ≥2R cellular rejection or ≥1 antibody-mediated rejection. The primary composite outcome was AR, graft dysfunction (left ventricular ejection fraction <50% and decrease by ≥10%), or death. RESULTS We included 148 patients (65 Black patients and 83 White patients), median age was 56 years and 30% female sex. The incidence of AR was higher in Black patients compared with White patients (43% versus 19%; P=0.002). Antibody-mediated rejection occurred predominantly in Black patients with a prevalence of 20% versus 2% (P<0.001). After transplant, Black patients had higher levels of dd-cfDNA, 0.09% (interquartile range, 0.001-0.30) compared with White patients, 0.05% (interquartile range, 0.001-0.23; P=0.003). Beyond 6 months, Black patients showed a persistent rise in dd-cfDNA with higher levels compared with White patients. Cell-free mitochondrial DNA was higher in Black patients (185 788 copies/mL; interquartile range, 101 252-422 133) compared with White patients (133 841 copies/mL; interquartile range, 75 346-337 990; P<0.001). The primary composite outcome occurred in 43% and 55% of Black patients at 1 and 2 years, compared with 23% and 27% in White patients, P<0.001. In a multivariable model, Black patient race (hazard ratio, 2.61 [95% CI, 1.35-5.04]; P=0.004) and %dd-cfDNA (hazard ratio, 1.15 [95% CI, 1.03-1.28]; P=0.010) were associated with the primary composite outcome. CONCLUSIONS Elevated dd-cfDNA and cell-free mitochondrial DNA after heart transplant may mechanistically be implicated in the higher incidence of AR and worse clinical outcomes in Black transplant recipients. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02423070.
Collapse
Affiliation(s)
- Palak Shah
- Heart Failure, Mechanical Circulatory Support & Transplant, Inova Heart and Vascular Institute, Falls Church VA
- Genomic Research Alliance for Transplantation (GRAfT), 10 Center Drive, 7S261, Bethesda Maryland, 20982
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT), 10 Center Drive, 7S261, Bethesda Maryland, 20982
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore MD
- Applied Precision Genomics, National Heart, Lung and Blood Institute, Bethesda MD
| | - Seiyon Lee
- Volgenau School of Engineering, George Mason University, Fairfax VA
| | - Temesgen E. Andargie
- Genomic Research Alliance for Transplantation (GRAfT), 10 Center Drive, 7S261, Bethesda Maryland, 20982
- Applied Precision Genomics, National Heart, Lung and Blood Institute, Bethesda MD
| | - Shashank S. Sinha
- Heart Failure, Mechanical Circulatory Support & Transplant, Inova Heart and Vascular Institute, Falls Church VA
| | - Hyesik Kong
- Applied Precision Genomics, National Heart, Lung and Blood Institute, Bethesda MD
| | - Lawrence Henry
- Heart Failure, Mechanical Circulatory Support & Transplant, Inova Heart and Vascular Institute, Falls Church VA
| | - Woojin Park
- Applied Precision Genomics, National Heart, Lung and Blood Institute, Bethesda MD
| | - Erick McNair
- Heart Failure, Mechanical Circulatory Support & Transplant, Inova Heart and Vascular Institute, Falls Church VA
| | - Inna Tchoukina
- The Pauley Heart Center, Virginia Commonwealth University, Richmond VA
| | - Keyur B. Shah
- The Pauley Heart Center, Virginia Commonwealth University, Richmond VA
| | - Samer S. Najjar
- Advanced Heart Failure Program, Medstar Heart and Vascular Institute, Washington Hospital Center, Washington DC
| | - Steven Hsu
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore MD
| | - Maria E. Rodrigo
- Advanced Heart Failure Program, Medstar Heart and Vascular Institute, Washington Hospital Center, Washington DC
| | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation (GRAfT), 10 Center Drive, 7S261, Bethesda Maryland, 20982
- Applied Precision Genomics, National Heart, Lung and Blood Institute, Bethesda MD
| | - Charles Marboe
- Department of Pathology, New York Presbyterian University Hospital of Cornell and Columbia, New York, New York, USA
| | | | - Hannah A. Valantine
- Genomic Research Alliance for Transplantation (GRAfT), 10 Center Drive, 7S261, Bethesda Maryland, 20982
- Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
25
|
Shi H, Yuan M, Cai J, Shi J, Li Y, Qian Q, Dong Z, Pan G, Zhu S, Wang W, Zhou J, Zhou X, Liu J. Exploring personalized treatment for cardiac graft rejection based on a four-archetype analysis model and bioinformatics analysis. Sci Rep 2024; 14:6529. [PMID: 38499711 PMCID: PMC10948767 DOI: 10.1038/s41598-024-57097-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/14/2024] [Indexed: 03/20/2024] Open
Abstract
Heart transplantation is the gold standard for treating patients with advanced heart failure. Although improvements in immunosuppressive therapies have significantly reduced the frequency of cardiac graft rejection, the incidences of T cell-mediated rejection (TCMR) and antibody-mediated rejection remain almost unchanged. A four-archetype analysis (4AA) model, developed by Philip F. Halloran, illustrated this problem well. It provided a new dimension to improve the accuracy of diagnoses and an independent system for recalibrating the histology guidelines. However, this model was based on the invasive method of endocardial biopsy, which undoubtedly increased the postoperative risk of heart transplant patients. Currently, little is known regarding the associated genes and specific functions of the different phenotypes. We performed bioinformatics analysis (using machine-learning methods and the WGCNA algorithm) to screen for hub-specific genes related to different phenotypes, based Gene Expression Omnibus accession number GSE124897. More immune cell infiltration was observed with the ABMR, TCMR, and injury phenotypes than with the stable phenotype. Hub-specific genes for each of the four archetypes were verified successfully using an external test set (accession number GSE2596). Logistic-regression models based on TCMR-specific hub genes and common hub genes were constructed with accurate diagnostic utility (area under the curve > 0.95). RELA, NFKB1, and SOX14 were identified as transcription factors important for TCMR/injury phenotypes and common genes, respectively. Additionally, 11 Food and Drug Administration-approved drugs were chosen from the DrugBank Database for each four-archetype model. Tyrosine kinase inhibitors may be a promising new option for transplant rejection treatment. KRAS signaling in cardiac transplant rejection is worth further investigation. Our results showed that heart transplant rejection subtypes can be accurately diagnosed by detecting expression of the corresponding specific genes, thereby enabling precise treatment or medication.
Collapse
Affiliation(s)
- Hongjie Shi
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Ming Yuan
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Jie Cai
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Jiajun Shi
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Yang Li
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Qiaofeng Qian
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Zhe Dong
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Gaofeng Pan
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Shaoping Zhu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Wei Wang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Jianliang Zhou
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Xianwu Zhou
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China.
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China.
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| |
Collapse
|
26
|
Goldberg JF, Mehta A, Bahniwal RK, Agbor-Enoh S, Shah P. A gentler approach to monitor for heart transplant rejection. Front Cardiovasc Med 2024; 11:1349376. [PMID: 38380175 PMCID: PMC10876874 DOI: 10.3389/fcvm.2024.1349376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
Despite developments in circulating biomarker and imaging technology in the assessment of cardiovascular disease, the surveillance and diagnosis of heart transplant rejection has continued to rely on histopathologic interpretation of the endomyocardial biopsy. Increasing evidence shows the utility of molecular evaluations, such as donor-specific antibodies and donor-derived cell-free DNA, as well as advanced imaging techniques, such as cardiac magnetic resonance imaging, in the assessment of rejection, resulting in the elimination of many surveillance endomyocardial biopsies. As non-invasive technologies in heart transplant rejection continue to evolve and are incorporated into practice, they may supplant endomyocardial biopsy even when rejection is suspected, allowing for more precise and expeditious rejection therapy. This review describes the current and near-future states for the evaluation of heart transplant rejection, both in the settings of rejection surveillance and rejection diagnosis. As biomarkers of rejection continue to evolve, rejection risk prediction may allow for a more personalized approach to immunosuppression.
Collapse
Affiliation(s)
- Jason F. Goldberg
- Department of Heart Failure and Transplantation, Inova Heart and Vascular Institute, Falls Church, VA, United States
- Department of Children's Cardiology, Inova L.J. Murphy Children’s Hospital, Falls Church, VA, United States
| | - Aditya Mehta
- Department of Heart Failure and Transplantation, Inova Heart and Vascular Institute, Falls Church, VA, United States
| | | | - Sean Agbor-Enoh
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, United States
| | - Palak Shah
- Department of Heart Failure and Transplantation, Inova Heart and Vascular Institute, Falls Church, VA, United States
| |
Collapse
|
27
|
Moayedi Y, Rodenas-Alesina E, Somerset E, Fan CPS, Henricksen E, Aleksova N, Billia F, Chih S, Ross HJ, Teuteberg JJ. Enhancing the Prediction of Cardiac Allograft Vasculopathy Using Intravascular Ultrasound and Machine Learning: A Proof of Concept. Circ Heart Fail 2024; 17:e011306. [PMID: 38314558 DOI: 10.1161/circheartfailure.123.011306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Cardiac allograft vasculopathy (CAV) is the leading cause of late graft dysfunction in heart transplantation. Building on previous unsupervised learning models, we sought to identify CAV clusters using serial maximal intimal thickness and baseline clinical risk factors to predict the development of early CAV. METHODS This is a single-center retrospective study including adult heart transplantation recipients. A latent class mixed-effects model was used to identify patient clusters with similar trajectories of maximal intimal thickness posttransplant and pretransplant covariates associated with each cluster. RESULTS Among 186 heart transplantation recipients, we identified 4 patient phenotypes: very low, low, moderate, and high risk. The 5-year risk (95% CI) of the International Society for Heart and Lung Transplantation-defined CAV in the high, moderate, low, and very low risk groups was 49.1% (35.2%-68.5%), 23.4% (13.3%-41.2%), 5.0% (1.3%-19.6%), and 0%, respectively. Only patients in the moderate to high risk cluster developed the International Society for Heart and Lung Transplantation CAV 2-3 at 5 years (P=0.02). Of the 4 groups, the low risk group had significantly younger female recipients, shorter ischemic time, and younger female donors compared with the high risk group. CONCLUSIONS We identified 4 clusters characterized by distinct maximal intimal thickness trajectories. These clusters were shown to discriminate against the development of angiographic CAV. This approach allows for the personalization of surveillance and CAV-directed treatment before the development of angiographically apparent disease.
Collapse
Affiliation(s)
- Yasbanoo Moayedi
- Ted Rogers Centre of Excellence in Heart Research (Y.M., E.R.-A., N.A., F.B., H.J.R.), Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada (Y.M., N.A., F.B., H.J.R.)
| | - Eduard Rodenas-Alesina
- Ted Rogers Centre of Excellence in Heart Research (Y.M., E.R.-A., N.A., F.B., H.J.R.), Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Emily Somerset
- Ted Rogers Computational Program, Centre of Excellence in Heart Function (E.S., C.P.S.F.), Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Chun Po S Fan
- Ted Rogers Computational Program, Centre of Excellence in Heart Function (E.S., C.P.S.F.), Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | | | - Natasha Aleksova
- Ted Rogers Centre of Excellence in Heart Research (Y.M., E.R.-A., N.A., F.B., H.J.R.), Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada (Y.M., N.A., F.B., H.J.R.)
| | - Filio Billia
- Ted Rogers Centre of Excellence in Heart Research (Y.M., E.R.-A., N.A., F.B., H.J.R.), Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada (Y.M., N.A., F.B., H.J.R.)
| | - Sharon Chih
- Ottawa Heart Institute, University of Ottawa, ON, Canada (S.C.)
| | - Heather J Ross
- Ted Rogers Centre of Excellence in Heart Research (Y.M., E.R.-A., N.A., F.B., H.J.R.), Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada (Y.M., N.A., F.B., H.J.R.)
| | - Jeffrey J Teuteberg
- Section of Heart Failure, Cardiac Transplant, and Mechanical Circulatory Support, Department of Medicine, Stanford University, CA (J.J.T.)
| |
Collapse
|
28
|
Shin M, Iyengar A, Helmers MR, Song C, Rekhtman D, Kelly JJ, Weingarten N, Patrick WL, Cevasco M. Non-inferior outcomes in lower urgency patients transplanted with extended criteria donor hearts. J Heart Lung Transplant 2024; 43:263-271. [PMID: 37778527 DOI: 10.1016/j.healun.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/07/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Recent work has suggested that outcomes among heart transplant patients listed at the lower-urgency (United Network for Organ Sharing Status 4 or 6) status may not be significantly impacted by donor comorbidities. The purpose of this study was to investigate outcomes of extended criteria donors (ECD) in lower versus higher urgency patients undergoing heart transplantation. METHODS The United Network for Organ Sharing (UNOS) database was queried for all adult patients undergoing heart transplantation from October 18, 2018 through December 31, 2021. Patients were stratified by degree of urgency (higher urgency: UNOS 1 or 2 vs lower urgency: UNOS 4 or 6) and receipt of ECD hearts, as defined by donor hearts failing to meet established acceptable use criteria. Outcomes were compared using propensity score matched cohorts. RESULTS Among 9,160 patients included, 2,320 (25.4%) were low urgency. ECD hearts were used in 35.5% of higher urgency (HU) patients and 39.2% of lower urgency (LU) patients. While ECD hearts had an impact on survival among high-urgency patients (p < 0.01), there was no difference in 1- and 2-year survival (p > 0.05) found among low urgency patients receiving ECD versus standard hearts. Neither ECDs nor individual ECD criteria were independently associated with mortality in low urgency patients (p > 0.05). CONCLUSIONS Post-transplant outcomes among low urgency patients are not adversely affected by receipt of ECD vs. standard hearts. Expanding the available donor pool by optimizing use of ECDs in this population may increase transplant frequency, decrease waitlist morbidity, and improve postoperative outcomes for the transplant community at large.
Collapse
Affiliation(s)
- Max Shin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amit Iyengar
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark R Helmers
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cindy Song
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Rekhtman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John J Kelly
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noah Weingarten
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - William L Patrick
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marisa Cevasco
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
29
|
Rodenas-Alesina E, Aleksova N, Stubbs M, Foroutan F, Kozuszko S, Posada JD, McDonald M, Moayedi Y, Ross H, Dipchand A. Cardiac allograft vasculopathy and survival in pediatric heart transplant recipients transitioned to adult care. J Heart Lung Transplant 2024; 43:229-237. [PMID: 37704160 DOI: 10.1016/j.healun.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Cardiac allograft vasculopathy (CAV) is an important cause of mortality after pediatric heart transplantation (HT) but there is a paucity of data regarding its incidence and impact on survival in pediatric recipients transitioned to adult care. METHODS We conducted a retrospective review of consecutive pediatric HT patients from 1989 to 2017 at the Hospital for Sick Children who transitioned to adult care at ≥18 years at Toronto General Hospital. We evaluated the incidence of International Society of Heart and Lung Transplantation CAV grade ≥1 using competing risk models. We assessed the association between all-cause mortality and CAV using Cox proportional hazards and used Kaplan Meier methods to evaluate all-cause mortality stratified by CAV and transplant era (1989-2001, 2002-2017). RESULTS Ninety-six patients were transitioned to adult care by January 2022, of which 53 underwent repeat coronary angiography as adults. CAV was newly diagnosed in 49% patients after transition to adult care. The overall incidence of CAV was 3.9 cases per 100 person-years. There was no difference in the adjusted incidence of CAV according to transplant era (subdistribution hazard ratios = 1.17, 95% confidence interval (CI) 0.54-2.66). CAV was associated with a higher risk of death in the early era (hazard ratio (HR) 10.29, 95% CI 2.16-49.96), but not in the recent era (HR 1.61, 95% 0.35-7.47). CONCLUSIONS There is a role for continued CAV surveillance after the transition to adult care. The implications of diagnosing CAV after the transition to adult care require further study, particularly because the risk of death in pediatric HT recipients diagnosed with CAV in the more recent era may be attenuated compared to the earlier HT era.
Collapse
Affiliation(s)
| | - Natasha Aleksova
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada; Women's College Hospital, Toronto, Ontario, Canada.
| | - Michael Stubbs
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Farid Foroutan
- Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Stella Kozuszko
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Juan Duero Posada
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Michael McDonald
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Yasbanoo Moayedi
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Heather Ross
- Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Anne Dipchand
- Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Chang A, Martin KA, Colvin M, Bellumkonda L. Role of ascorbic acid in cardiac allograft vasculopathy. Clin Transplant 2023; 37:e15153. [PMID: 37792313 DOI: 10.1111/ctr.15153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/04/2023] [Accepted: 09/22/2023] [Indexed: 10/05/2023]
Abstract
PURPOSE OF THE REVIEW Cardiac allograft vasculopathy (CAV) is a progressive fibroproliferative disease which occurs after heart transplantation and is associated with significant long-term morbidity and mortality. Currently available strategies including statins, mammalian target of rapamycin (mTOR) inhibitors, and revascularization, have limited overall effectiveness in treating this pathology once the disease process is established. mTOR inhibitors, while effective when used early in the disease process, are not well tolerated, and hence not routinely used in post-transplant care. RECENT DATA Recent work on rodent models have given us a novel mechanistic understanding of effects of ascorbic acid in preventing CAV. TET methyl cytosine dioxygenase2 (TET2) reduces vascular smooth muscle cell (VSMC) apoptosis and intimal thickening. TET2 is repressed by interferon γ (IFNγ) in the setting of CAV. Ascorbic acid has been shown to promote TET2 activity and attenuate allograft vasculopathy in animal models and CAV progression in a small clinical trial. SUMMARY CAV remains a challenging disease process and needs better preventative strategies. Ascorbic acid improves endothelial dysfunction, reduces reactive oxygen species, and prevents development of intimal hyperplasia by preventing smooth muscle cell apoptosis and hyperproliferation. Further large-scale randomized control studies of ascorbic acid are needed to establish the role in routine post-transplant management.
Collapse
Affiliation(s)
- Alyssa Chang
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathleen A Martin
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Monica Colvin
- Division of Cardiology, Department of Medicine, Yale University, New Haven, Connecticut, USA
| | - Lavanya Bellumkonda
- Division of Cardiology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Suarez-Pierre A, Iguidbashian J, Kirsch MJ, Cotton JL, Quinn C, Fullerton DA, Reece TB, Hoffman JRH, Cleveland JC, Rove JY. Importance of social vulnerability on long-term outcomes after heart transplantation. Am J Transplant 2023; 23:1580-1589. [PMID: 37414250 DOI: 10.1016/j.ajt.2023.06.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023]
Abstract
The relationship between social determinants of health and outcomes after heart transplantation has not been examined. The social vulnerability index (SVI) uses United States census data to determine the social vulnerability of every census tract based on 15 factors. This retrospective study seeks to examine the impact of SVI on outcomes after heart transplantation. Adult heart recipients who received a graft between 2012 and 2021 were stratified into SVI percentiles of <75% and SVI of ≥75%. The primary endpoint was survival. The median SVI was 48% (interquartile range: 30%-67%) among 23 700 recipients. One-year survival was similar between groups (91.4 vs 90.7%, log-rank P = .169); however, 5-year survival was lower among individuals living in vulnerable communities (74.8% vs 80.0%, P < .001). This finding persisted despite risk adjustment for other factors associated with mortality (survival time ratio 0.819, 95% confidence interval: 0.755-0.890, P < .001). The incidences of 5-year hospital readmission (81.4% vs 75.4%, P < .001) and graft rejection (40.3% vs 35.7%, P = .004) were higher among individuals living in vulnerable communities. Individuals living in vulnerable communities may be at increased risk of mortality after heart transplantation. These findings suggest there is an opportunity to focus on these recipients undergoing heart transplantation to improve survival.
Collapse
Affiliation(s)
- Alejandro Suarez-Pierre
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA.
| | - John Iguidbashian
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael J Kirsch
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jake L Cotton
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Christopher Quinn
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - David A Fullerton
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Thomas Brett Reece
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jordan R H Hoffman
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Joseph C Cleveland
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jessica Y Rove
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
32
|
DeFilippis EM, Nikolova A, Holzhauser L, Khush KK. Understanding and Investigating Sex-Based Differences in Heart Transplantation: A Call to Action. JACC. HEART FAILURE 2023; 11:1181-1188. [PMID: 37589612 DOI: 10.1016/j.jchf.2023.06.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 08/18/2023]
Abstract
Women represent only about 25% of heart transplant recipients annually. Although the number of women living with advanced heart failure remains unknown, epidemiologic research suggests that more women should be receiving advanced heart failure therapies. Sex differences in risk factors, presentation, response to pharmacotherapy, and outcomes in heart failure have been well described. Yet, less is known about sex differences in heart transplant candidate selection, waitlist management, donor selection, perioperative considerations, and post-transplant management and outcomes. The purpose of this review was to summarize the existing published reports related to sex differences in heart transplantation, highlighting areas in which sex-based considerations are well described and supported by available evidence, and emphasizing topics that require further study.
Collapse
Affiliation(s)
- Ersilia M DeFilippis
- Division of Cardiology, Columbia University Irving Medical Center, New York, New York, USA
| | - Andriana Nikolova
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Luise Holzhauser
- Division of Cardiovascular Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kiran K Khush
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California, USA.
| |
Collapse
|
33
|
Kondziella C, Fluschnik N, Weimann J, Schrage B, Becher PM, Memenga F, Bernhardt AM, Blankenberg S, Reichenspurner H, Kirchhof P, Schnabel RB, Magnussen C. Sex differences in clinical characteristics and outcomes in patients undergoing heart transplantation. ESC Heart Fail 2023; 10:2596-2606. [PMID: 37339937 PMCID: PMC10375178 DOI: 10.1002/ehf2.14413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/04/2023] [Accepted: 05/02/2023] [Indexed: 06/22/2023] Open
Abstract
AIMS Whether sex affects selection for and outcomes after heart transplantation (HTx) remains unclear. We aimed to show sex differences in pre-transplant characteristics and outcomes after HTx. METHODS AND RESULTS From 1995 to 2019, 49 200 HTx recipients were prospectively enrolled in the Organ Procurement and Transplantation Network. Logistic regression models were used to evaluate clinical characteristics by sex. Multivariable Cox regression models were fitted to assess sex differences in all-cause mortality, cardiovascular mortality, graft failure, cardiac allograft vasculopathy (CAV), and malignancy. In 49 200 patients (median age 55 years, interquartile range 46-62; 24.6% women), 49 732 events occurred during a median follow-up of 8.1 years. Men were older than women, had more often ischaemic cardiomyopathy (odds ratio [OR] 3.26, 95% confidence interval [CI] 3.11-3.42; P < 0.001), and a higher burden of cardiovascular risk factors, whereas women had less malignancies (OR 0.47, CI 0.44-0.51; P < 0.001). Men were more often treated in intensive care unit (OR 1.24, CI 1.12-1.37; P < 0.001) with a higher need for ventilatory (OR 1.24, CI 1.17-1.32; P < 0.001) or VAD (OR 1.53, CI 1.45-1.63; P < 0.001) support. After multivariable adjustment, men had a higher risk for CAV (hazard ratio [HR] 1.21, CI 1.13-1.29; P < 0.001) and malignancy (HR 1.80, CI 1.62-2.00; P < 0.001). There were no differences in all-cause mortality, cardiovascular mortality, and graft failure between sexes. CONCLUSIONS In this US transplant registry, men and women differed in pre-transplant characteristics. Male sex was independently associated with incident CAV and malignancy even after multivariable adjustment. Our results underline the need for better personalized post-HTx management and care.
Collapse
Affiliation(s)
- Christoph Kondziella
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Nina Fluschnik
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Jessica Weimann
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Benedikt Schrage
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Peter Moritz Becher
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Felix Memenga
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Alexander M. Bernhardt
- Department of Cardiovascular Surgery, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Stefan Blankenberg
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Hermann Reichenspurner
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
- Department of Cardiovascular Surgery, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Paulus Kirchhof
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
- Institute of Cardiovascular SciencesUniversity of BirminghamBirminghamUK
| | - Renate B. Schnabel
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| | - Christina Magnussen
- Department of Cardiology, University Heart & Vascular Center HamburgUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- German Center of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/LuebeckHamburgGermany
| |
Collapse
|
34
|
Bonnet G, Coutance G, Aubert O, Waldmann V, Raynaud M, Asselin A, Bories MC, Guillemain R, Bruneval P, Varnous S, Leprince P, Achouch P, Marijon E, Loupy A, Jouven X. Sudden cardiac death after heart transplantation: a population-based study. Europace 2023; 25:euad126. [PMID: 37208303 PMCID: PMC10198773 DOI: 10.1093/europace/euad126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
AIMS The epidemiology of sudden cardiac death (SCD) after heart transplantation (HTx) remains imprecisely described. We aimed to assess the incidence and determinants of SCD in a large cohort of HTx recipients, compared with the general population. METHODS AND RESULTS Consecutive HTx recipients (n = 1246, 2 centres) transplanted between 2004 and 2016 were included. We prospectively assessed clinical, biological, pathologic, and functional parameters. SCD was centrally adjudicated. We compared the SCD incidence beyond the first year post-transplant in this cohort with that observed in the general population of the same geographic area (registry carried out by the same group of investigators; n = 19 706 SCD). We performed a competing risk multivariate Cox model to identify variables associated with SCD. The annual incidence of SCD was 12.5 per 1,000 person-years [95% confidence interval (CI), 9.7-15.9] in the HTx recipients cohort compared with 0.54 per 1,000 person-years (95% CI, 0.53-0.55) in the general population (P < 0.001). The risk of SCD was markedly elevated among the youngest HTx recipients with standardized mortality ratios for SCD up to 837 for recipients ≤30 years. Beyond the first year, SCD was the leading cause of death. Five variables were independently associated with SCD: older donor age (P = 0.003), younger recipient age (P = 0.001) and ethnicity (P = 0.034), pre-existing donor-specific antibodies (P = 0.009), and last left ventricular ejection fraction (P = 0.048). CONCLUSION HTx recipients, particularly the youngest, were at very high risk of SCD compared with the general population. The consideration of specific risk factors may help identify high-risk subgroups.
Collapse
Affiliation(s)
- Guillaume Bonnet
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
- UMCV, Haut-Lévêque Hospital, University Hospital of Bordeaux, 33600 Pessac, France
| | - Guillaume Coutance
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
- Department of Cardiac and Thoracic Surgery, Cardiology Institute, Pitié-Salpeêtrière Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University Medical School, Paris, France
| | - Olivier Aubert
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Victor Waldmann
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
- Cardiology and Heart Transplant department, European Georges Pompidou Hospital, Assistance Publique—Hôpitaux de Paris, Rue Leblanc, 75015 Paris, France
| | - Marc Raynaud
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
| | - Anouk Asselin
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
| | - Marie-Cécile Bories
- Cardiology and Heart Transplant department, European Georges Pompidou Hospital, Assistance Publique—Hôpitaux de Paris, Rue Leblanc, 75015 Paris, France
| | - Romain Guillemain
- Cardiology and Heart Transplant department, European Georges Pompidou Hospital, Assistance Publique—Hôpitaux de Paris, Rue Leblanc, 75015 Paris, France
| | - Patrick Bruneval
- Pathology Department, Georges Pompidou Hospital, Assistance Publique—Hôpitaux de Paris. Université de Paris, Paris, France
| | - Shaida Varnous
- Department of Cardiac and Thoracic Surgery, Cardiology Institute, Pitié-Salpeêtrière Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University Medical School, Paris, France
- INSERM, UMRS-1166, iCAN, Institute of Cardiometabolism and Nutrition, Hôpital de la Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Université Pierre et Marie Curie, Paris, France
| | - Pascal Leprince
- Department of Cardiac and Thoracic Surgery, Cardiology Institute, Pitié-Salpeêtrière Hospital, Assistance Publique des Hôpitaux de Paris (AP-HP), Sorbonne University Medical School, Paris, France
- INSERM, UMRS-1166, iCAN, Institute of Cardiometabolism and Nutrition, Hôpital de la Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Université Pierre et Marie Curie, Paris, France
| | - Paul Achouch
- Cardiology and Heart Transplant department, European Georges Pompidou Hospital, Assistance Publique—Hôpitaux de Paris, Rue Leblanc, 75015 Paris, France
| | - Eloi Marijon
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
- Cardiology and Heart Transplant department, European Georges Pompidou Hospital, Assistance Publique—Hôpitaux de Paris, Rue Leblanc, 75015 Paris, France
| | - Alexandre Loupy
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
- Kidney Transplant Department, Necker Hospital, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Xavier Jouven
- Université de Paris, Paris Cardiovascular Research Center (PARCC), Paris Translational Research Center for Organ Transplantation, INSERM, UMR-S970, 75015 Paris, France
- Cardiology and Heart Transplant department, European Georges Pompidou Hospital, Assistance Publique—Hôpitaux de Paris, Rue Leblanc, 75015 Paris, France
| |
Collapse
|
35
|
Franco-Acevedo A, Comes J, Mack JJ, Valenzuela NM. New insights into maladaptive vascular responses to donor specific HLA antibodies in organ transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1146040. [PMID: 38993843 PMCID: PMC11235244 DOI: 10.3389/frtra.2023.1146040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/03/2023] [Indexed: 07/13/2024]
Abstract
Transplant vasculopathy (TV) causes thickening of donor blood vessels in transplanted organs, and is a significant cause of graft loss and mortality in allograft recipients. It is known that patients with repeated acute rejection and/or donor specific antibodies are predisposed to TV. Nevertheless, the exact molecular mechanisms by which alloimmune injury culminates in this disease have not been fully delineated. As a result of this incomplete knowledge, there is currently a lack of effective therapies for this disease. The immediate intracellular signaling and the acute effects elicited by anti-donor HLA antibodies are well-described and continuing to be revealed in deeper detail. Further, advances in rejection diagnostics, including intragraft gene expression, provide clues to the inflammatory changes within allografts. However, mechanisms linking these events with long-term outcomes, particularly the maladaptive vascular remodeling seen in transplant vasculopathy, are still being delineated. New evidence demonstrates alterations in non-coding RNA profiles and the occurrence of endothelial to mesenchymal transition (EndMT) during acute antibody-mediated graft injury. EndMT is also readily apparent in numerous settings of non-transplant intimal hyperplasia, and lessons can be learned from advances in those fields. This review will provide an update on these recent developments and remaining questions in our understanding of HLA antibody-induced vascular damage, framed within a broader consideration of manifestations and implications across transplanted organ types.
Collapse
Affiliation(s)
- Adriana Franco-Acevedo
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| | - Johanna Comes
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Julia J Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA, United States
| | - Nicole M Valenzuela
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, United States
| |
Collapse
|
36
|
Cho I, Kim WD, Kim S, Ko KY, Seong Y, Kim DY, Seo J, Shim CY, Ha JW, Mori M, Gupta A, You SC, Hong GR, Krumholz HM. Reclassification of moderate aortic stenosis based on data-driven phenotyping of hemodynamic progression. Sci Rep 2023; 13:6694. [PMID: 37095171 PMCID: PMC10125992 DOI: 10.1038/s41598-023-33683-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 04/17/2023] [Indexed: 04/26/2023] Open
Abstract
The management and follow-up of moderate aortic stenosis (AS) lacks consensus as the progression patterns are not well understood. This study aimed to identify the hemodynamic progression of AS, and associated risk factors and outcomes. We included patients with moderate AS with at least three transthoracic echocardiography (TTE) studies performed between 2010 and 2021. Latent class trajectory modeling was used to classify AS groups with distinctive hemodynamic trajectories, which were determined by serial systolic mean pressure gradient (MPG) measurements. Outcomes were defined as all-cause mortality and aortic valve replacement (AVR). A total of 686 patients with 3093 TTE studies were included in the analysis. Latent class model identified two distinct AS trajectory groups based on their MPG: a slow progression group (44.6%) and a rapid progression group (55.4%). Initial MPG was significantly higher in the rapid progression group (28.2 ± 5.6 mmHg vs. 22.9 ± 2.8 mmHg, P < 0.001). The prevalence of atrial fibrillation was higher in the slow progression group; there was no significant between-group difference in the prevalence of other comorbidities. The rapid progression group had a significantly higher AVR rate (HR 3.4 [2.4-4.8], P < 0.001); there was no between-group difference in mortality (HR 0.7 [0.5-1.0]; P = 0.079). Leveraging longitudinal echocardiographic data, we identified two distinct groups of patients with moderate AS: slow and rapid progression. A higher initial MPG (≥ 24 mmHg) was associated with more rapid progression of AS and higher rates of AVR, thus indicating the predictive value of MPG in management of the disease.
Collapse
Affiliation(s)
- Iksung Cho
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - William D Kim
- Chung-Ang University College of Medicine, Seoul, Korea
| | - Subin Kim
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea
- Institute for Innovation in Digital Healthcare, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Kyu-Yong Ko
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Yeonchan Seong
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Dae-Young Kim
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jiwon Seo
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Chi Young Shim
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jong-Won Ha
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Makoto Mori
- Division of Cardiac Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
| | - Aakriti Gupta
- Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Seng Chan You
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Korea.
- Institute for Innovation in Digital Healthcare, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| | - Geu-Ru Hong
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| | - Harlan M Krumholz
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
37
|
Anwar T, Sinnett-Smith J, Jin YP, Reed EF, Rozengurt E. Lipophilic Statins Inhibit YAP Nuclear Localization, Coactivator Activity, and Migration in Response to Ligation of HLA Class I Molecules in Endothelial Cells: Role of YAP Multisite Phosphorylation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1134-1145. [PMID: 36881871 PMCID: PMC10073314 DOI: 10.4049/jimmunol.2200568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 02/10/2023] [Indexed: 03/09/2023]
Abstract
Solid-organ transplant recipients exhibiting HLA donor-specific Abs are at risk for graft loss due to chronic Ab-mediated rejection. HLA Abs bind HLA molecules expressed on the surface of endothelial cells (ECs) and induce intracellular signaling pathways, including the activation of the transcriptional coactivator yes-associated protein (YAP). In this study, we examined the impact of lipid-lowering drugs of the statin family on YAP localization, multisite phosphorylation, and transcriptional activity in human ECs. Exposure of sparse cultures of ECs to cerivastatin or simvastatin induced striking relocalization of YAP from the nucleus to the cytoplasm and inhibited the expression of the YAP/TEA domain DNA-binding transcription factor-regulated genes connective tissue growth factor and cysteine-rich angiogenic inducer 61. In dense cultures of ECs, statins prevented YAP nuclear import and expression of connective tissue growth factor and cysteine-rich angiogenic inducer 61 stimulated by the mAb W6/32 that binds HLA class I. Exposure of ECs to either cerivastatin or simvastatin completely blocked the migration of ECs stimulated by ligation of HLA class I. Exogenously supplied mevalonic acid or geranylgeraniol reversed the inhibitory effects of statins on YAP localization either in low-density ECs or high-density ECs challenged with W6/32. Mechanistically, cerivastatin increased the phosphorylation of YAP at Ser127, blunted the assembly of actin stress fiber, and inhibited YAP phosphorylation at Tyr357 in ECs. Using mutant YAP, we substantiated that YAP phosphorylation at Tyr357 is critical for YAP activation. Collectively, our results indicate that statins restrain YAP activity in EC models, thus providing a plausible mechanism underlying their beneficial effects in solid-organ transplant recipients.
Collapse
Affiliation(s)
- Tarique Anwar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - James Sinnett-Smith
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- VA Greater Los Angeles Health System
| | - Yi-Ping Jin
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- VA Greater Los Angeles Health System
| |
Collapse
|
38
|
First- Versus New-Generation Drug-Eluting Stents in Patients With Heart Transplant With Cardiac Allograft Vasculopathy. Am J Cardiol 2023; 188:7-14. [PMID: 36446228 DOI: 10.1016/j.amjcard.2022.10.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/11/2022] [Accepted: 10/30/2022] [Indexed: 11/27/2022]
Abstract
Although several studies have previously reported on the efficacy of percutaneous coronary intervention (PCI) with first-generation drug-eluting stents (DES) in heart transplant patients with cardiac allograft vasculopathy, few data regarding new-generation DES are currently available. We sought to compare the efficacy of new-generation versus first-generation DES in 90 consecutive patients with heart transplant (113 de novo coronary lesions) who underwent urgent or elective PCI with first-generation (28 patients) or new-generation (62 patients) DES. For each patient, the severity of cardiac allograft vasculopathy and postprocedural extent of revascularization were quantified calculating baseline and residual SYNTAX score, respectively. The primary end point was a composite of major adverse cardiac events-myocardial infarction, cardiovascular death, or target vessel revascularization-at 3 years. Overall, the median baseline SYNTAX score was 8 (5 to 15), and a total number of stents per patient of 1.6 ± 0.9 was implanted. Post-PCI residual SYNTAX score was 1.5 (0 to 4), with 13 patients having a score >8. At 3 years, the Kaplan-Meier estimate of freedom from major adverse cardiac events was 64%, with no differences between first-generation and new-generation DES groups (log-rank test p = 0.269). Nevertheless, patients treated with new-generation DES experienced a lower rate of target vessel revascularization (15% vs 31%, log-rank test p = 0.058). In the multivariate Cox regression analysis, a post-PCI residual SYNTAX score >8 (hazard ratio 2.37, confidence interval 0.98 to 5.73, p = 0.054) was identified as an independent predictor of the primary end point.
Collapse
|
39
|
Masoud AG, Lin J, Zhu LF, Tao K, Ness NW, Kassiri Z, Moore RB, Vanhaesebroeck B, West L, Anderson CC, Oudit GY, Murray AG. Endothelial phosphoinositide 3-kinase-β inactivation confers protection from immune-mediated vascular injury. Am J Transplant 2023; 23:202-213. [PMID: 36804130 DOI: 10.1016/j.ajt.2022.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/03/2022] [Accepted: 11/02/2022] [Indexed: 01/06/2023]
Abstract
Heart transplant and recipient survival are limited by immune cell-mediated injury of the graft vasculature. We examined the role of the phosphoinositide 3-kinase-β (PI3Kβ) isoform in endothelial cells (EC) during coronary vascular immune injury and repair in mice. In minor histocompatibility-antigen mismatched allogeneic heart grafts, a robust immune response was mounted to each wild-type, PI3Kβ inhibitor-treated, or endothelial-selective PI3Kβ knockout (ECβKO) graft transplanted to wild-type recipients. However, microvascular EC loss and progressive occlusive vasculopathy only developed in control, but not PI3Kβ-inactivated hearts. We observed a delay in inflammatory cell infiltration of the ECβKO grafts, particularly in the coronary arteries. Surprisingly, this was accompanied by an impaired display of proinflammatory chemokine and adhesion molecules by the ECβKO ECs. In vitro, tumor necrosis factor α-stimulated endothelial ICAM1 and VCAM1 expression was blocked by PI3Kβ inhibition or RNA interference. Selective PI3Kβ inhibition also blocked tumor necrosis factor α-stimulated degradation of inhibitor of nuclear factor kappa Bα and nuclear translocation of nuclear factor kappa B p65 in EC. These data identify PI3Kβ as a therapeutic target to reduce vascular inflammation and injury.
Collapse
Affiliation(s)
- Andrew G Masoud
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Alberta Transplant Institute, Edmonton, Alberta, Canada
| | - Jiaxin Lin
- Alberta Transplant Institute, Edmonton, Alberta, Canada; Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Lin F Zhu
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Kesheng Tao
- Alberta Transplant Institute, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Nathan W Ness
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Alberta Transplant Institute, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Ronald B Moore
- Alberta Transplant Institute, Edmonton, Alberta, Canada; Department of Surgery, University of Alberta, Edmonton, Alberta, Canada; Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | | | - Lori West
- Alberta Transplant Institute, Edmonton, Alberta, Canada; Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Colin C Anderson
- Alberta Transplant Institute, Edmonton, Alberta, Canada; Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; UCL Cancer Institute, University College London, London, England, UK; Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Allan G Murray
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Alberta Transplant Institute, Edmonton, Alberta, Canada.
| |
Collapse
|
40
|
The Molecular Microscope Diagnostic System: Assessment of Rejection and Injury in Heart Transplant Biopsies. Transplantation 2023; 107:27-44. [PMID: 36508644 DOI: 10.1097/tp.0000000000004323] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review describes the development of the Molecular Microscope Diagnostic System (MMDx) for heart transplant endomyocardial biopsies (EMBs). MMDx-Heart uses microarrays to measure biopsy-based gene expression and ensembles of machine learning algorithms to interpret the results and compare each new biopsy to a large reference set of earlier biopsies. MMDx assesses T cell-mediated rejection (TCMR), antibody-mediated rejection (AMR), recent parenchymal injury, and atrophy-fibrosis, continually "learning" from new biopsies. Rejection-associated transcripts mapped in kidney transplants and experimental systems were used to identify TCMR, AMR, and recent injury-induced inflammation. Rejection and injury emerged as gradients of intensity, rather than binary classes. AMR was one-third donor-specific antibody (DSA)-negative, and many EMBs first considered to have no rejection displayed minor AMR-like changes, with increased probability of DSA positivity and subtle inflammation. Rejection-associated transcript-based algorithms now classify EMBs as "Normal," "Minor AMR changes," "AMR," "possible AMR," "TCMR," "possible TCMR," and "recent injury." Additionally, MMDx uses injury-associated transcript sets to assess the degree of parenchymal injury and atrophy-fibrosis in every biopsy and study the effect of rejection on the parenchyma. TCMR directly injures the parenchyma whereas AMR usually induces microcirculation stress but relatively little initial parenchymal damage, although slowly inducing parenchymal atrophy-fibrosis. Function (left ventricular ejection fraction) and short-term risk of failure are strongly determined by parenchymal injury. These discoveries can guide molecular diagnostic applications, either as a central MMDx system or adapted to other platforms. MMDx can also help calibrate noninvasive blood-based biomarkers to avoid unnecessary biopsies and monitor response to therapy.
Collapse
|
41
|
Clinical recommendations for posttransplant assessment of anti-HLA (Human Leukocyte Antigen) donor-specific antibodies: A Sensitization in Transplantation: Assessment of Risk consensus document. Am J Transplant 2023; 23:115-132. [PMID: 36695614 DOI: 10.1016/j.ajt.2022.11.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/29/2022] [Accepted: 11/04/2022] [Indexed: 01/13/2023]
Abstract
Although anti-HLA (Human Leukocyte Antigen) donor-specific antibodies (DSAs) are commonly measured in clinical practice and their relationship with transplant outcome is well established, clinical recommendations for anti-HLA antibody assessment are sparse. Supported by a careful and critical review of the current literature performed by the Sensitization in Transplantation: Assessment of Risk 2022 working group, this consensus report provides clinical practice recommendations in kidney, heart, lung, and liver transplantation based on expert assessment of quality and strength of evidence. The recommendations address 3 major clinical problems in transplantation and include guidance regarding posttransplant DSA assessment and application to diagnostics, prognostics, and therapeutics: (1) the clinical implications of positive posttransplant DSA detection according to DSA status (ie, preformed or de novo), (2) the relevance of posttransplant DSA assessment for precision diagnosis of antibody-mediated rejection and for treatment management, and (3) the relevance of posttransplant DSA for allograft prognosis and risk stratification. This consensus report also highlights gaps in current knowledge and provides directions for clinical investigations and trials in the future that will further refine the clinical utility of posttransplant DSA assessment, leading to improved transplant management and patient care.
Collapse
|
42
|
Louis K, Lefaucheur C. DSA in solid organ transplantation: is it a matter of specificity, amount, or functional characteristics? Curr Opin Organ Transplant 2022; 27:392-398. [PMID: 35881421 DOI: 10.1097/mot.0000000000001006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW The present review describes the clinical relevance of human leukocyte antigen (HLA) donor-specific antibodies (HLA-DSAs) as biomarkers of alloimmunity and summarizes recent improvements in their characterization that provide insights into immune risk assessment, precision diagnosis, and prognostication in transplantation. RECENT FINDINGS Recent studies have addressed the clinical utility of HLA-DSAs as biomarkers for immune risk assessment in pretransplant and peritransplant, diagnosis and treatment evaluation of antibody-mediated rejection, immune monitoring posttransplant, and risk stratification. SUMMARY HLA-DSAs have proved to be the most advanced immune biomarkers in solid organ transplantation in terms of analytical validity, clinical validity and clinical utility. Recent studies are integrating multiple HLA-DSA characteristics including antibody specificity, HLA class, quantity, immunoglobulin G subclass, and complement-binding capacity to improve risk assessment peritransplant, diagnosis and treatment evaluation of antibody-mediated rejection, immune monitoring posttransplant, and transplant prognosis evaluation. In addition, integration of HLA-DSAs to clinical, functional and histological transplant parameters has further consolidated the utility of HLA-DSAs as robust biomarkers and allows to build new tools for monitoring, precision diagnosis, and risk stratification for individual patients. However, prospective and randomized-controlled studies addressing the clinical benefit and cost-effectiveness of HLA-DSA-based monitoring and patient management strategies are required to demonstrate that the use of HLA-DSAs as biomarkers can improve current clinical practice and transplant outcomes.
Collapse
Affiliation(s)
- Kevin Louis
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris
- Human Immunology and Immunopathology, Université de Paris
| | - Carmen Lefaucheur
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris
- Paris Translational Research Center for Organ Transplantation, Institut national de la santé et de la recherche médicale UMR-S970, Université de Paris, Paris, France
| |
Collapse
|
43
|
Assessing the Relationship Between Molecular Rejection and Parenchymal Injury in Heart Transplant Biopsies. Transplantation 2022; 106:2205-2216. [PMID: 35968995 DOI: 10.1097/tp.0000000000004231] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND The INTERHEART study (ClinicalTrials.gov #NCT02670408) used genome-wide microarrays to detect rejection in endomyocardial biopsies; however, many heart transplants with no rejection have late dysfunction and impaired survival. We used the microarray measurements to develop a molecular classification of parenchymal injury. METHODS In 1320 endomyocardial biopsies from 645 patients previously studied for rejection-associated transcripts, we measured the expression of 10 injury-induced transcript sets: 5 induced by recent injury; 2 reflecting macrophage infiltration; 2 normal heart transcript sets; and immunoglobulin transcripts, which correlate with time. We used archetypal clustering to assign injury groups. RESULTS Injury transcript sets correlated with impaired function. Archetypal clustering based on the expression of injury transcript sets assigned each biopsy to 1 of 5 injury groups: 87 Severe-injury, 221 Late-injury, and 3 with lesser degrees of injury, 376 No-injury, 526 Mild-injury, and 110 Moderate-injury. Severe-injury had extensive loss of normal transcripts (dedifferentiation) and increase in macrophage and injury-induced transcripts. Late-injury was characterized by high immunoglobulin transcript expression. In Severe- and Late-injury, function was depressed, and short-term graft failure was increased, even in hearts with no rejection. T cell-mediated rejection almost always had parenchymal injury, and 85% had Severe- or Late-injury. In contrast, early antibody-mediated rejection (ABMR) had little injury, but late ABMR often had the Late-injury state. CONCLUSION Characterizing heart transplants for their injury state provides new understanding of dysfunction and outcomes and demonstrates the differential impact of T cell-mediated rejection versus ABMR on the parenchyma. Slow deterioration from ABMR emerges as a major contributor to late dysfunction.
Collapse
|
44
|
Ko K, Cho I, Kim S, Seong Y, Kim D, Seo JW, You SC, Shim CY, Hong G, Ha J. Identification of Distinct Subgroups in Moderately Severe Rheumatic Mitral Stenosis Using Data-Driven Phenotyping of Longitudinal Hemodynamic Progression. J Am Heart Assoc 2022; 11:e026375. [PMID: 35904199 PMCID: PMC9375495 DOI: 10.1161/jaha.121.026375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022]
Abstract
Background Rheumatic mitral stenosis is a significant cause of valvular heart disease. Pulmonary arterial systolic pressure (PASP) reflects the hemodynamic consequences of mitral stenosis and is used to determine treatment strategies. However, PASP progression and expected outcomes based on PASP changes in patients with moderately severe mitral stenosis remain unclear. Methods and Results A total of 436 patients with moderately severe rheumatic mitral stenosis (valve area 1.0-1.5 cm2) were enrolled. Composite outcomes included all-cause mortality and hospitalization for heart failure. Data-driven phenotyping identified 2 distinct trajectory groups based on PASP progression: rapid (8.7%) and slow (91.3%). Patients in the rapid progression group were older and had more diabetes and atrial fibrillation than those in the slow progression group (all P<0.05). The initial mean diastolic pressure gradient and PASP were higher in the rapid progression group than in the slow progression group (6.2±2.4 mm Hg versus 5.1±2.0 mm Hg [P=0.001] and 42.3±13.3 mm Hg versus 33.0±9.2 mm Hg [P<0.001], respectively). The rapid progression group had a poorer event-free survival rate than the slow progression group (log-rank P<0.001). Rapid PASP progression was a significant risk factor for composite outcomes even after adjusting for comorbidities (hazard ratio, 3.08 [95% CI, 1.68-5.64]; P<0.001). Multivariate regression analysis revealed that PASP >40 mm Hg was independently associated with allocation to the rapid progression group (odds ratio, 4.95 [95% CI, 2.08-11.99]; P<0.001). Conclusions Rapid PASP progression was associated with a higher risk of the composite outcomes. The main independent predictor for rapid progression group allocation was initial PASP >40 mm Hg.
Collapse
Affiliation(s)
- Kyu‐Yong Ko
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Iksung Cho
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Subin Kim
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Yeonchan Seong
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Dae‐Young Kim
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Ji Won Seo
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Seng Chan You
- Department of Preventive MedicineYonsei University College of MedicineSeoulKorea
| | - Chi Young Shim
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Geu‐Ru Hong
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Jong‐Won Ha
- Division of Cardiology, Severance HospitalYonsei University College of MedicineSeoulKorea
| |
Collapse
|
45
|
Tsuda H, Dvorina N, Keslar KS, Nevarez-Mejia J, Valenzuela NM, Reed EF, Fairchild RL, Baldwin WM. Molecular Signature of Antibody-Mediated Chronic Vasculopathy in Heart Allografts in a Novel Mouse Model. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1053-1065. [PMID: 35490714 PMCID: PMC9253905 DOI: 10.1016/j.ajpath.2022.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 04/23/2023]
Abstract
Cardiac allograft vasculopathy (CAV) limits the long-term success of heart transplants. Generation of donor-specific antibodies (DSAs) is associated with increased incidence of CAV clinically, but mechanisms underlying development of this pathology remain poorly understood. Major histocompatibility complex-mismatched A/J cardiac allografts in B6.CCR5-/- recipients have been reported to undergo acute rejection with little T-cell infiltration, but intense deposition of C4d in large vessels and capillaries of the graft accompanied by high titers of DSA. This model was modified to investigate mechanisms of antibody-mediated CAV by transplanting A/J hearts to B6.CCR5-/- CD8-/- mice that were treated with low doses of anti-CD4 monoclonal antibody to decrease T-cell-mediated graft injury and promote antibody-mediated injury. Although the mild inhibition of CD4 T cells extended allograft survival, the grafts developed CAV with intense C4d deposition and macrophage infiltration by 14 days after transplantation. Development of CAV correlated with recipient DSA titers. Transcriptomic analysis of microdissected allograft arteries identified the Notch ligand Dll4 as the most elevated transcript in CAV, associated with high versus low titers of DSA. More importantly, these analyses revealed a differential expression of transcripts in the CAV lesions compared with the matched apical tissue that lacks large arteries. In conclusion, these findings report a novel model of antibody-mediated CAV with the potential to facilitate further understanding of the molecular mechanisms promoting development of CAV.
Collapse
Affiliation(s)
- Hidetoshi Tsuda
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Karen S Keslar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jessica Nevarez-Mejia
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Nicole M Valenzuela
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Elaine F Reed
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Robert L Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| | - William M Baldwin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
46
|
Coutance G, Patel JK. Seeing Old Landscapes With New Eyes: A Voyage Into the Endomyocardial Biopsy to Improve Risk Stratification After Heart Transplant Using Computational Analysis. Circulation 2022; 145:1578-1580. [PMID: 35605035 DOI: 10.1161/circulationaha.122.059933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Guillaume Coutance
- Department of Cardiac and Thoracic Surgery, Cardiology Institute, Pitié Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Sorbonne University Medical School, France (G.C.).,University of Paris, INSERM UMR 970, Paris Translational Research Centre for Organ Transplantation, France (G.C.)
| | - Jignesh K Patel
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (J.K.P.)
| |
Collapse
|
47
|
Peyster EG, Janowczyk A, Swamidoss A, Kethireddy S, Feldman MD, Margulies KB. Computational Analysis of Routine Biopsies Improves Diagnosis and Prediction of Cardiac Allograft Vasculopathy. Circulation 2022; 145:1563-1577. [PMID: 35405081 DOI: 10.1161/circulationaha.121.058459] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Cardiac allograft vasculopathy (CAV) is a leading cause of morbidity and mortality for heart transplant recipients. While clinical risk factors for CAV have been established, no personalized prognostic test exists to confidently identify patients at high vs. low risk of developing aggressive CAV. The aim of this investigation was to leverage computational methods for analyzing digital pathology images from routine endomyocardial biopsies (EMB) to develop a precision medicine tool for predicting CAV years before overt clinical presentation. Methods: Clinical data from 1-year post-transplant was collected on 302 transplant recipients from the University of Pennsylvania, including 53 'early CAV' patients and 249 'no-CAV' controls. This data was used to generate a 'clinical model' (ClinCAV-Pr) for predicting future CAV development. From this cohort, n=183 archived EMBs were collected for CD31 and modified trichrome staining and then digitally scanned. These included 1-year post-transplant EMBs from 50 'early CAV' patients and 82 no-CAV patients, as well as 51 EMBs from 'disease control' patients obtained at the time of definitive coronary angiography confirming CAV. Using biologically-inspired, hand-crafted features extracted from digitized EMBs, quantitative histologic models for differentiating no-CAV from disease controls (HistoCAV-Dx), and for predicting future CAV from 1-year post-transplant EMBs were developed (HistoCAV-Pr). The performance of histologic and clinical models for predicting future CAV (i.e. HistoCAV-Pr and ClinCAV-Pr, respectively) were compared in a held-out validation set, before being combined to assess the added predictive value of an integrated predictive model (iCAV-Pr). Results: ClinCAV-Pr achieved modest performance on the independent test set, with area under the receiver operating curve (AUROC) of 0.70. The HistoCAV-Dx model for diagnosing CAV achieved excellent discrimination, with an AUROC of 0.91, while HistoCAV-Pr model for predicting CAV achieved good performance with an AUROC of 0.80. The integrated iCAV-Pr model achieved excellent predictive performance, with an AUROC of 0.93 on the held-out test set. Conclusions: Prediction of future CAV development is greatly improved by incorporation of computationally extracted histologic features. These results suggest morphologic details contained within regularly obtained biopsy tissue have the potential to enhance precision and personalization of treatment plans for post-heart transplant patients.
Collapse
Affiliation(s)
- Eliot G Peyster
- Cardiovascular Research Institute (E.G.P., K.B.M.), University of Pennsylvania, Philadelphia
| | - Andrew Janowczyk
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH (A.J., A.S., S.K.)
- Department of Oncology, Lausanne University Hospital and Lausanne University, Switzerland (A.J.)
| | - Abigail Swamidoss
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH (A.J., A.S., S.K.)
| | - Samhith Kethireddy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH (A.J., A.S., S.K.)
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine (M.D.F.), University of Pennsylvania, Philadelphia
| | - Kenneth B Margulies
- Cardiovascular Research Institute (E.G.P., K.B.M.), University of Pennsylvania, Philadelphia
| |
Collapse
|
48
|
Baran DA, Mohammed A, Macdonald P, Copeland H. Heart Transplant Donor Selection: Recent Insights. CURRENT TRANSPLANTATION REPORTS 2022. [DOI: 10.1007/s40472-022-00355-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
49
|
Cardiac Allograft Vasculopathy in Pediatric Heart Transplant Recipients Does Early-Onset Portend a Worse Prognosis? J Heart Lung Transplant 2022; 41:578-588. [DOI: 10.1016/j.healun.2022.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/05/2021] [Accepted: 01/08/2022] [Indexed: 11/19/2022] Open
|
50
|
Aleksova N, Umar F, Bernick J, Mielniczuk LM, Ross HJ, Chih S. Low-Density Lipoprotein Cholesterol Level Trends and the Development of Cardiac Allograft Vasculopathy After Heart Transplantation. CJC Open 2021; 3:1453-1462. [PMID: 34993457 PMCID: PMC8712546 DOI: 10.1016/j.cjco.2021.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/12/2021] [Indexed: 11/26/2022] Open
Abstract
Background Unlike the relationship with atherosclerotic coronary artery disease, that between low-density lipoprotein cholesterol (LDL-C) and cardiac allograft vasculopathy (CAV) is unclear. Our objectives were to characterize lipid profiles early after heart transplantation (HT) and evaluate the relationship between early LDL-C and the development of CAV. Methods We retrospectively reviewed consecutive adults who underwent HT at 2 centres during the time period 2010-2018. The primary outcome was the incidence of angiographic CAV. The relationship between LDL-C and CAV was assessed using Cox proportional hazards and logistic regression models adjusted a priori for clinically important covariates, including recipient and donor age, recipient sex, ischemic time, and pre-HT diabetes. Results A total of 386 patients followed for a median (range) of 4.4 (2.8-6.8) years were included. LDL-C at baseline (2.11 ± 0.86 mmol/L) and 1 year after HT (2.20 ± 0.88 mmol/L) was similar (P = 0.21), but it was lower at the end of follow-up (1.89 ± 0.74 mmol/L, P < 0.01). Of 309 patients who underwent angiography, 54% had CAV. The risk of CAV did not vary according to baseline, 1-year, or change from baseline to 1-year LDL-C. The odds of CAV at 1 year were equally likely across LDL-C values (adjusted odds ratio 1.00, 95% confidence interval: 0.61-1.63 for baseline, and adjusted odds ratio 1.25, 95% confidence interval: 0.74-2.10 for 1-year LDL-C). Conclusions No association was identified between early LDL-C and the development of CAV. Our findings do not support targeting a specific LDL-C for patients who do not otherwise meet criteria for guideline-recommended LDL-C target levels. Randomized studies are warranted to determine if lipid-lowering to a specific LDL-C target level modifies the risk of CAV.
Collapse
|