1
|
Vernooij RW, Michael M, Colombijn JM, Owers DS, Webster AC, Strippoli GF, Hodson EM. Pre-emptive treatment for cytomegalovirus viraemia to prevent cytomegalovirus disease in solid organ transplant recipients. Cochrane Database Syst Rev 2025; 1:CD005133. [PMID: 39807668 PMCID: PMC11729901 DOI: 10.1002/14651858.cd005133.pub4] [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] [Indexed: 01/16/2025]
Abstract
BACKGROUND Cytomegalovirus (CMV) is a significant cause of morbidity and death in solid organ transplant recipients. Pre-emptive treatment of patients with CMV viraemia using antiviral agents has been suggested as an alternative to routine prophylaxis to prevent CMV disease. This is an update of a Cochrane review first published in 2006 and updated in 2013. OBJECTIVES To determine the benefits and harms of pre-emptive treatment of CMV viraemia to prevent CMV disease and death (any cause) and the indirect effects of CMV infection (acute rejection, graft loss, opportunistic infections) in solid organ transplant recipients. SEARCH METHODS The Cochrane Kidney and Transplant Register of Studies was searched up to 17 December 2024 using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal, and ClinicalTrials.gov. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing pre-emptive treatment with placebo, no specific treatment, or antiviral prophylaxis in solid organ transplant recipients. DATA COLLECTION AND ANALYSIS Two authors independently assessed the eligibility of the identified studies, assessed the risk of bias, and extracted all data. Results were expressed as risk ratio (RR) and 95% confidence intervals (CI) for dichotomous outcomes. Statistical analyses were performed using a random-effects model. The certainty of evidence was assessed per outcome using the Grades of Recommendation, Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS In this update, we have included seven new studies, bringing the total number of included studies to 22 (1883 participants). Of these, seven investigated pre-emptive treatment versus placebo or standard care, 12 looked at pre-emptive treatment versus antiviral prophylaxis, one study investigated oral versus intravenous pre-emptive treatment, one investigated pre-emptive valganciclovir versus pre-emptive ganciclovir, and one investigated letermovir 40 mg twice/day versus 80 mg once/day. Studies were conducted in Australia, Brazil, the Czech Republic, Germany, Italy, Japan, Norway, Spain, South Korea, and the USA. Organ transplant recipients included kidney, liver, heart, lung, and kidney-pancreas. Thirteen studies were single-centre studies, six were multicentre, and three were unknown. The number of participants ranged from 12 to 296. Overall, selection bias was unclear (55%); performance, detection and attrition bias were high (91%, 63% and 95%, respectively), and reporting bias was low (55%). Compared with placebo or standard care, pre-emptive treatment probably reduces the risk of CMV disease (7 studies, 315 participants: RR 0.29, 95% CI 0.11 to 0.80; I2 = 54%; moderate-certainty evidence) but may result in little or no difference in death (any cause) (3 studies, 176 participants: RR 1.23, 95% CI 0.35 to 4.30; I2 = 0%; low-certainty evidence). Pre-emptive treatment may result in little or no difference in CMV organ involvement, CMV-associated symptoms, acute rejection, graft loss, other infections or leucopenia. Compared to prophylaxis, pre-emptive treatment may make little or no difference to the risk of developing CMV disease (11 studies, 1322 participants: RR 0.97, 95% CI 0.47 to 2.01; I2 = 54%; low-certainty evidence) and probably makes little or no difference to death (any cause) (9 studies, 1098 participants: RR 0.95, 95% CI 0.60 to 1.52; I2 = 0%; moderate-certainty evidence). Pre-emptive treatment may increase the risk of CMV infection (8 studies, 867 participants: RR 1.97, 95% CI 1.48 to 2.61; I2 = 66%; low-certainty evidence). The risk of leucopenia (7 studies, 869 participants: RR 0.57, 95% CI 0.38 to 0.87; I2 = 33%; moderate-certainty evidence) and neutropenia (5 studies, 859 participants: RR 0.63, 95% CI 0.44 to 0.90; I2 = 0% moderate certainty evidence) probably decreases with pre-emptive therapy. There may be little or no difference in the risks of acute rejection, graft loss, and infections other than CMV. Single studies were identified for comparisons between different pre-emptive treatments: 1) oral ganciclovir versus IV ganciclovir; 2) valganciclovir versus ganciclovir; 3) 40 mg twice/day versus 80 mg once/day. No differences between these treatment modalities in terms of CMV disease, death (any cause), or adverse events were identified. AUTHORS' CONCLUSIONS In this review, we have included seven new studies, yet the available evidence is overall of low certainty and the conclusions remain similar to the previous version of this review. Pre-emptive treatment probably reduces the risk of CMV disease compared with placebo or standard care. There were no clear differences between pre-emptive treatment and prophylaxis to prevent CMV disease or reduce the risk of death (any cause). The risk of CMV infection may be higher for patients receiving pre-emptive therapy, but the risk of adverse events, such as leucopenia, is probably lower.
Collapse
Affiliation(s)
- Robin Wm Vernooij
- Department of Nephrology and Hypertension and Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mini Michael
- Division of Pediatric Nephrology, Department of Pediatrics, Texas Children's Hospital/Baylor College of Medicine, Houston, TX, USA
| | - Julia Mt Colombijn
- Department of Nephrology and Hypertension and Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daniel S Owers
- Department of Critical Care, The Canberra Hospital, Garran, Australia
| | - Angela C Webster
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Westmead Applied Research Centre, The University of Sydney at Westmead, Westmead, Australia
- Department of Transplant and Renal Medicine, Westmead Hospital, Westmead, Australia
| | - Giovanni Fm Strippoli
- Department of Precision and Regenerative Medicine and Ionian Area (Dimepre-J), University of Bari, Bari, Italy
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Elisabeth M Hodson
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| |
Collapse
|
2
|
Gupta C, Mundan NG, Das S, Jawed A, Dar SA, Dailah HG. Cytomegalovirus Infections in Hematopoietic Stem Cell Transplant: Moving Beyond Molecular Diagnostics to Immunodiagnostics. Diagnostics (Basel) 2024; 14:2523. [PMID: 39594189 PMCID: PMC11592488 DOI: 10.3390/diagnostics14222523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/25/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Human CMV, regularly reactivated by simple triggers, results in asymptomatic viral shedding, powerful cellular immune responses, and memory inflation. Immunocompetent individuals benefit from a robust immune response, which aids in viral management without causing clinically significant illness; however, immunodeficient individuals are always at a higher risk of CMV reactivation and disease. Hematopoietic stem cell transplant (HSCT) recipients are consistently at higher risk of CMV reactivation and clinically significant CMV illness due to primary disease, immunosuppression, and graft vs. host disease. Early recovery of CMV-CMI responses may mitigate effects of viral reactivation in HSCT recipients. Immune reconstitution following transplantation occurs spontaneously and is mediated initially by donor-derived T cells, followed by clonal growth of T cells produced from graft progenitors. CMV-specific immune reconstitution post-transplant is related to spontaneous clearance of CMV reactivation and may eliminate the need for prophylactic or pre-emptive medication, making it a potential predictive marker for monitoring CMV reactivation. This review highlights current thoughts and therapeutic options for CMV reactivation in HSCT, with focus on CMV immune reconstitution and post-HSCT monitoring. Immune monitoring aids in risk stratification of transplant recipients who may progress from CMV reactivation to clinically significant CMV infection. Implementing this approach in clinical practice reduces the need for periodic viral surveillance and antiviral therapy in recipients who have a high CMV-CMI and thus may experience self-limited reactivation. Therefore, in the age of precision medicine, it is critical to incorporate CMV-specific cellular immune surveillance into conventional procedures and algorithms for the management of transplant recipients.
Collapse
Affiliation(s)
- Chhavi Gupta
- Department of Infectious Diseases, Yashoda Super Speciality Hospital, Ghaziabad 201001, India
| | - Netto George Mundan
- Department of Infectious Diseases, Government Medical College, Kottayam 686008, India
| | - Shukla Das
- Department of Microbiology, University College of Medical Sciences and GTB Hospital (University of Delhi), Delhi 110095, India
| | - Arshad Jawed
- College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia (S.A.D.)
| | - Sajad Ahmad Dar
- College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia (S.A.D.)
| | - Hamad Ghaleb Dailah
- College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia (S.A.D.)
| |
Collapse
|
3
|
Schleiss MR, Crooks CM, Karthigeyan KP, Kruc RM, Otero CE, Wang HY(S, Permar SR, Plotkin SA, Gautam R. Proceedings of the Conference "CMV Vaccine Development-How Close Are We?" (27-28 September 2023). Vaccines (Basel) 2024; 12:1231. [PMID: 39591134 PMCID: PMC11598149 DOI: 10.3390/vaccines12111231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 11/28/2024] Open
Abstract
Congenital cytomegalovirus (cCMV) is the most common infectious cause of disability in children, including sensorineural hearing loss. There is interest in developing a pre-conception vaccine that could confer protective immunity on a woman of child-bearing age, hence resulting in a reduced cCMV disease burden. Other populations, including solid organ transplant (SOT) and hematopoietic stem cell transplant (HSCT) patients, could also benefit from CMV vaccination. To review and discuss vaccines that are in clinical development, a workshop, sponsored by the National Institutes of Health (NIH) and the National Institute of Allergy and Infectious Diseases (NIAID), was empaneled. At this workshop, correlates of protective immunity against CMV, epidemiologic features of CMV transmission, and vaccine platforms in development were reviewed. Representatives from academia, pharma, and the NIH engaged in discussion on the current state-of-the-art in CMV vaccinology. A summary of the presentations from this is provided in this report.
Collapse
Affiliation(s)
- Mark R. Schleiss
- Division of Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Chelsea M. Crooks
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Krithika P. Karthigeyan
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Rebecca M. Kruc
- Department of Pediatrics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Claire E. Otero
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Hsuan-Yuan (Sherry) Wang
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, 1300 York Ave Box 65, New York, NY 10065, USA;
| | - Stanley A. Plotkin
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Rajeev Gautam
- Program Officer at Virology Branch, Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fisher’s Lane, Rockville, MD 20892, USA;
| |
Collapse
|
4
|
Scherger SJ, Molina KC, Palestine AG, Pecen PE, Bajrovic V. Cytomegalovirus Retinitis in the Modern Era of Solid Organ Transplantation. Transplant Proc 2024; 56:1696-1701. [PMID: 39147617 PMCID: PMC11401753 DOI: 10.1016/j.transproceed.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Cytomegalovirus retinitis (CMVR) is a well-described complication of CMV disease in immunocompromised hosts. While robust data exists for CMVR in patients with acquired immunodeficiency syndrome (AIDS), the incidence and risk factors for CMVR in solid organ transplant recipients (SOTR) with CMV viremia are less defined. METHODS We performed a retrospective cohort study of SOTR who had CMV viremia and underwent routine ophthalmologic examination between 1/1/2018 and 3/16/2022. Univariate statistics were performed to evaluate risk factors for development of CMVR. RESULTS Overall, 38 patients were included, primarily kidney (78.9%), heart (7.9%), and liver (7.9%) transplant recipients. Five patients (13.2%) developed CMVR during the study period. CMVR was diagnosed an average 281 days after index transplantation, 84 days from the most recent rejection episode, and 69 days from onset of viremia. Only 1 patient (20%) had symptoms at the time of CMVR diagnosis. CMVR was associated with preceding allograft rejection as well as transplanted organ type. CONCLUSION While CMV tissue disease more commonly manifests in other organs, CMVR occurred relatively frequently in this group of high-risk SOTR with CMV viremia. As most of the patients in our study did not have ocular symptoms at the time of diagnosis, routine ophthalmologic screening should be considered in SOTR with CMV viremia.
Collapse
Affiliation(s)
- Sias J Scherger
- Department of Medicine, Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Kyle C Molina
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, CO, USA; Department of Pharmacy, Scripps Health, La Jolla, CA, USA
| | - Alan G Palestine
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, USA
| | - Paula E Pecen
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, USA
| | - Valida Bajrovic
- Department of Medicine, Icahn School of Medicine at Mt. Sinai, Division of Infectious Diseases, New York, NY, USA
| |
Collapse
|
5
|
Berg T, Aehling NF, Bruns T, Welker MW, Weismüller T, Trebicka J, Tacke F, Strnad P, Sterneck M, Settmacher U, Seehofer D, Schott E, Schnitzbauer AA, Schmidt HH, Schlitt HJ, Pratschke J, Pascher A, Neumann U, Manekeller S, Lammert F, Klein I, Kirchner G, Guba M, Glanemann M, Engelmann C, Canbay AE, Braun F, Berg CP, Bechstein WO, Becker T, Trautwein C. S2k-Leitlinie Lebertransplantation der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) und der Deutschen Gesellschaft für Allgemein- und Viszeralchirurgie (DGAV). ZEITSCHRIFT FUR GASTROENTEROLOGIE 2024; 62:1397-1573. [PMID: 39250961 DOI: 10.1055/a-2255-7246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Affiliation(s)
- Thomas Berg
- Bereich Hepatologie, Medizinischen Klinik II, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Niklas F Aehling
- Bereich Hepatologie, Medizinischen Klinik II, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Tony Bruns
- Medizinische Klinik III, Universitätsklinikum Aachen, Aachen, Deutschland
| | - Martin-Walter Welker
- Medizinische Klinik I Gastroent., Hepat., Pneum., Endokrin. Universitätsklinikum Frankfurt, Frankfurt, Deutschland
| | - Tobias Weismüller
- Klinik für Innere Medizin - Gastroenterologie und Hepatologie, Vivantes Humboldt-Klinikum, Berlin, Deutschland
| | - Jonel Trebicka
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Deutschland
| | - Frank Tacke
- Charité - Universitätsmedizin Berlin, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Campus Virchow-Klinikum (CVK) und Campus Charité Mitte (CCM), Berlin, Deutschland
| | - Pavel Strnad
- Medizinische Klinik III, Universitätsklinikum Aachen, Aachen, Deutschland
| | - Martina Sterneck
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Hamburg, Hamburg, Deutschland
| | - Utz Settmacher
- Klinik für Allgemein-, Viszeral- und Gefäßchirurgie, Universitätsklinikum Jena, Jena, Deutschland
| | - Daniel Seehofer
- Klinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Eckart Schott
- Klinik für Innere Medizin II - Gastroenterologie, Hepatologie und Diabetolgie, Helios Klinikum Emil von Behring, Berlin, Deutschland
| | | | - Hartmut H Schmidt
- Klinik für Gastroenterologie und Hepatologie, Universitätsklinikum Essen, Essen, Deutschland
| | - Hans J Schlitt
- Klinik und Poliklinik für Chirurgie, Universitätsklinikum Regensburg, Regensburg, Deutschland
| | - Johann Pratschke
- Chirurgische Klinik, Charité Campus Virchow-Klinikum - Universitätsmedizin Berlin, Berlin, Deutschland
| | - Andreas Pascher
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Universitätsklinikum Münster, Münster, Deutschland
| | - Ulf Neumann
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Universitätsklinikum Essen, Essen, Deutschland
| | - Steffen Manekeller
- Klinik und Poliklinik für Allgemein-, Viszeral-, Thorax- und Gefäßchirurgie, Universitätsklinikum Bonn, Bonn, Deutschland
| | - Frank Lammert
- Medizinische Hochschule Hannover (MHH), Hannover, Deutschland
| | - Ingo Klein
- Chirurgische Klinik I, Universitätsklinikum Würzburg, Würzburg, Deutschland
| | - Gabriele Kirchner
- Klinik und Poliklinik für Chirurgie, Universitätsklinikum Regensburg und Innere Medizin I, Caritaskrankenhaus St. Josef Regensburg, Regensburg, Deutschland
| | - Markus Guba
- Klinik für Allgemeine, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Universitätsklinikum München, München, Deutschland
| | - Matthias Glanemann
- Klinik für Allgemeine, Viszeral-, Gefäß- und Kinderchirurgie, Universitätsklinikum des Saarlandes, Homburg, Deutschland
| | - Cornelius Engelmann
- Charité - Universitätsmedizin Berlin, Medizinische Klinik m. S. Hepatologie und Gastroenterologie, Campus Virchow-Klinikum (CVK) und Campus Charité Mitte (CCM), Berlin, Deutschland
| | - Ali E Canbay
- Medizinische Klinik, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Deutschland
| | - Felix Braun
- Klinik für Allgemeine Chirurgie, Viszeral-, Thorax-, Transplantations- und Kinderchirurgie, Universitätsklinikum Schlewswig-Holstein, Kiel, Deutschland
| | - Christoph P Berg
- Innere Medizin I Gastroenterologie, Hepatologie, Infektiologie, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - Wolf O Bechstein
- Klinik für Allgemein- und Viszeralchirurgie, Universitätsklinikum Frankfurt, Frankfurt, Deutschland
| | - Thomas Becker
- Klinik für Allgemeine Chirurgie, Viszeral-, Thorax-, Transplantations- und Kinderchirurgie, Universitätsklinikum Schlewswig-Holstein, Kiel, Deutschland
| | | |
Collapse
|
6
|
Puget L, Node J, Caël B, Bamoulid J, Coaquette A, Prétet JL, Lepiller Q. Urinary cytomegalovirus excretion: The unresolved issues. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:755-761. [PMID: 38492661 DOI: 10.1016/j.pharma.2024.03.004] [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: 02/12/2024] [Revised: 03/06/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Cytomegalovirus (CMV) excretion in urine is frequently observed in clinical practice. However, the specific circumstances and pathophysiological mechanisms underlying this shedding remain largely unknown. Here, we address some of the key questions regarding urinary CMV excretion, focusing on new hypotheses raised by recent advances in the field. Cellular origins of CMV shedding, clinical contexts of occurrence, systemic spread of the virus versus compartmentalization in the urinary tract, and clinical impact are successively discussed.
Collapse
Affiliation(s)
- Line Puget
- Laboratoire de virologie, CHU de Besançon, Besançon, France
| | - Juliette Node
- Laboratoire de virologie, CHU de Besançon, Besançon, France
| | - Blandine Caël
- Laboratoire d'auto-immunité et d'allergologie, CHU Besançon, Besançon, France
| | - Jamal Bamoulid
- Service de néphrologie, CHU de Besançon, Besançon, France
| | - Alain Coaquette
- Laboratoire d'auto-immunité et d'allergologie, CHU Besançon, Besançon, France
| | - Jean-Luc Prétet
- Laboratoire de biologie cellulaire, CHU de Besançon, Besançon, France; UMR 6249 Laboratoire chrono-environnement, CNRS-UFC, Besançon, France
| | - Quentin Lepiller
- Laboratoire de virologie, CHU de Besançon, Besançon, France; UMR 6249 Laboratoire chrono-environnement, CNRS-UFC, Besançon, France.
| |
Collapse
|
7
|
Bharti R, Calabrese DR. Innate and adaptive effector immune drivers of cytomegalovirus disease in lung transplantation: a double-edged sword. FRONTIERS IN TRANSPLANTATION 2024; 3:1388393. [PMID: 38993763 PMCID: PMC11235306 DOI: 10.3389/frtra.2024.1388393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/24/2024] [Indexed: 07/13/2024]
Abstract
Up to 90% of the global population has been infected with cytomegalovirus (CMV), a herpesvirus that remains latent for the lifetime of the host and drives immune dysregulation. CMV is a critical risk factor for poor outcomes after solid organ transplant, though lung transplant recipients (LTR) carry the highest risk of CMV infection, and CMV-associated comorbidities compared to recipients of other solid organ transplants. Despite potent antivirals, CMV remains a significant driver of chronic lung allograft dysfunction (CLAD), re-transplantation, and death. Moreover, the extended utilization of CMV antiviral prophylaxis is not without adverse effects, often necessitating treatment discontinuation. Thus, there is a critical need to understand the immune response to CMV after lung transplantation. This review identifies key elements of each arm of the CMV immune response and highlights implications for lung allograft tolerance and injury. Specific attention is paid to cellular subsets of adaptive and innate immune cells that are important in the lung during CMV infection and reactivation. The concept of heterologous immune responses is reviewed in depth, including how they form and how they may drive tissue- and allograft-specific immunity. Other important objectives of this review are to detail the emerging role of NK cells in CMV-related outcomes, in addition to discussing perturbations in CMV immune function stemming from pre-existing lung disease. Finally, this review identifies potential mechanisms whereby CMV-directed treatments may alter the cellular immune response within the allograft.
Collapse
Affiliation(s)
- Reena Bharti
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Daniel R. Calabrese
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States
| |
Collapse
|
8
|
Kapoor H, Bi C, Kroll MH, Salm AE, Dominguez EA. Burden and frequency of viral testing of kidney and non-kidney transplant recipients. Microbiol Spectr 2024; 12:e0357523. [PMID: 38709030 PMCID: PMC11237713 DOI: 10.1128/spectrum.03575-23] [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: 10/06/2023] [Accepted: 04/15/2024] [Indexed: 05/07/2024] Open
Abstract
Transplant patients are at risk of infections due to long-term immunosuppression contributing to morbidity and mortality in this population. Post-transplant testing guidelines were established to monitor and guide therapeutic interventions in transplant recipients. We hypothesize that there are gaps in adherence to the recommended frequency of laboratory testing in post-transplant patients. We analyzed national reference laboratory data to compare viral post-transplant infection (PTI) testing frequency with their respective published guidelines to understand patient uptake and compliance. We evaluated the ordering patterns, positivity rates, and frequency of molecular infectious disease tests (MIDTs). We included 345 patients with International Classification of Diseases (ICD)-10 codes for transplant (Z940-Z942, Z944, Z9481, Z9483, Z9484) with at least two tests (within 7 days) in January 2019 and at least one test in December 2020 to find patients in the post-transplant period. We analyzed two cohorts: kidney transplant recipients (KTRs; 40%) and non-KTR (60%) then followed them longitudinally for the study period. In KTR cohort, high-to-low proportion of ordered MIDT was blood BK virus (bBKV) followed by cytomegalovirus (CMV); in non-KTR cohort, CMV was followed by Epstein-Barr virus (EBV). KTR cohort positivity was highest for urine BK virus (uBKV; 58%) followed by EBV (46%), bBKV (40%), and CMV (31%). Non-KTR cohort positivity was highest for uBKV (64%), EBV (51%), CMV (30%), bBKV (8%), and adenovirus (7%). All patients were tested at progressively longer intervals from the date of the first post-transplant ICD-10-coded test. More than 40% of the KTR cohort were tested less frequently for EBV and bBKV, and more than 20% of the non-KTR cohort were tested for EBV less frequently than published guidelines 4 months after transplant. Despite regular testing, the results of MIDT testing for KTR and non-KTR patients in the post-transplant period are not aligned with published guidelines.IMPORTANCEGuidance for post-transplant infectious disease testing is established, however, for certain infections it allows for clinician discretion. This leads to transplant center policies developing their own testing/surveillance strategies based on their specific transplant patient population (kidney, stem cell, etc.). The Organ Procurement and Transplant Network (OPTN) has developed a strategic plan to improve and standardize the transplant process in the US to improve outcomes of living donors and recipients. Publishing national reference lab data on the testing frequency and its alignment with the recommended guidelines for post-transplant infectious diseases can inform patient uptake and compliance for these strategic OPTN efforts.
Collapse
Affiliation(s)
- Hema Kapoor
- Infectious Diseases/Immunology, Quest Diagnostics, Secaucus, New Jersey, USA
| | - Caixia Bi
- Medical Informatics, Biostatistics and Outcomes Research Group, Quest Diagnostics, Secaucus, New Jersey, USA
| | - Martin H. Kroll
- Analytics and Statistical Applications, Quest Diagnostics, Secaucus, New Jersey, USA
| | - Ann E. Salm
- Infectious Diseases/Immunology, Quest Diagnostics, Secaucus, New Jersey, USA
| | - Edward A. Dominguez
- Organ Transplant Infectious Disease, Methodist Transplant Specialists, Dallas, Texas, USA
| |
Collapse
|
9
|
Vernooij RW, Michael M, Ladhani M, Webster AC, Strippoli GF, Craig JC, Hodson EM. Antiviral medications for preventing cytomegalovirus disease in solid organ transplant recipients. Cochrane Database Syst Rev 2024; 5:CD003774. [PMID: 38700045 PMCID: PMC11066972 DOI: 10.1002/14651858.cd003774.pub5] [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] [Indexed: 05/05/2024]
Abstract
BACKGROUND The risk of cytomegalovirus (CMV) infection in solid organ transplant recipients has resulted in the frequent use of prophylaxis to prevent the clinical syndrome associated with CMV infection. This is an update of a review first published in 2005 and updated in 2008 and 2013. OBJECTIVES To determine the benefits and harms of antiviral medications to prevent CMV disease and all-cause death in solid organ transplant recipients. SEARCH METHODS We contacted the information specialist and searched the Cochrane Kidney and Transplant Register of Studies up to 5 February 2024 using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Registry Platform (ICTRP) Search Portal, and ClinicalTrials.gov. SELECTION CRITERIA We included randomised controlled trials (RCTs) and quasi-RCTs comparing antiviral medications with placebo or no treatment, comparing different antiviral medications or different regimens of the same antiviral medications for CMV prophylaxis in recipients of any solid organ transplant. Studies examining pre-emptive therapy for CMV infection are studied in a separate review and were excluded from this review. DATA COLLECTION AND ANALYSIS Two authors independently assessed study eligibility, risk of bias and extracted data. Summary estimates of effect were obtained using a random-effects model, and results were expressed as risk ratios (RR) and their 95% confidence intervals (CI) for dichotomous outcomes and mean difference (MD) and 95% CI for continuous outcomes. Confidence in the evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS This 2024 update found four new studies, bringing the total number of included studies to 41 (5054 participants). The risk of bias was high or unclear across most studies, with a low risk of bias for sequence generation (12), allocation concealment (12), blinding (11) and selective outcome reporting (9) in fewer studies. There is high-certainty evidence that prophylaxis with aciclovir, ganciclovir or valaciclovir compared with placebo or no treatment is more effective in preventing CMV disease (19 studies: RR 0.42, 95% CI 0.34 to 0.52), all-cause death (17 studies: RR 0.63, 95% CI 0.43 to 0.92), and CMV infection (17 studies: RR 0.61, 95% CI 0.48 to 0.77). There is moderate-certainty evidence that prophylaxis probably reduces death from CMV disease (7 studies: RR 0.26, 95% CI 0.08 to 0.78). Prophylaxis reduces the risk of herpes simplex and herpes zoster disease, bacterial and protozoal infections but probably makes little to no difference to fungal infection, acute rejection or graft loss. No apparent differences in adverse events with aciclovir, ganciclovir or valaciclovir compared with placebo or no treatment were found. There is high certainty evidence that ganciclovir, when compared with aciclovir, is more effective in preventing CMV disease (7 studies: RR 0.37, 95% CI 0.23 to 0.60). There may be little to no difference in any outcome between valganciclovir and IV ganciclovir compared with oral ganciclovir (low certainty evidence). The efficacy and adverse effects of valganciclovir or ganciclovir were probably no different to valaciclovir in three studies (moderate certainty evidence). There is moderate certainty evidence that extended duration prophylaxis probably reduces the risk of CMV disease compared with three months of therapy (2 studies: RR 0.20, 95% CI 0.12 to 0.35), with probably little to no difference in rates of adverse events. Low certainty evidence suggests that 450 mg/day valganciclovir compared with 900 mg/day valganciclovir results in little to no difference in all-cause death, CMV infection, acute rejection, and graft loss (no information on adverse events). Maribavir may increase CMV infection compared with ganciclovir (1 study: RR 1.34, 95% CI: 1.10 to 1.65; moderate certainty evidence); however, little to no difference between the two treatments were found for CMV disease, all-cause death, acute rejection, and adverse events at six months (low certainty evidence). AUTHORS' CONCLUSIONS Prophylaxis with antiviral medications reduces CMV disease and CMV-associated death, compared with placebo or no treatment, in solid organ transplant recipients. These data support the continued routine use of antiviral prophylaxis in CMV-positive recipients and CMV-negative recipients of CMV-positive organ transplants.
Collapse
Affiliation(s)
- Robin Wm Vernooij
- Department of Nephrology and Hypertension and Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mini Michael
- Division of Pediatric Nephrology, Baylor College of Medicine, Houston, TX, USA
| | - Maleeka Ladhani
- Nephrology, Lyell McEwin Hospital, Elizabeth Vale, Australia
| | - Angela C Webster
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Westmead Applied Research Centre, The University of Sydney at Westmead, Westmead, Australia
- Centre for Transplant and Renal Medicine, Westmead Millennium Institute, The University of Sydney at Westmead, Westmead, Australia
| | - Giovanni Fm Strippoli
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Jonathan C Craig
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Elisabeth M Hodson
- Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| |
Collapse
|
10
|
Dettori M, Riccardi N, Canetti D, Antonello RM, Piana AF, Palmieri A, Castiglia P, Azara AA, Masia MD, Porcu A, Ginesu GC, Cossu ML, Conti M, Pirina P, Fois A, Maida I, Madeddu G, Babudieri S, Saderi L, Sotgiu G. Infections in lung transplanted patients: A review. Pulmonology 2024; 30:287-304. [PMID: 35710714 DOI: 10.1016/j.pulmoe.2022.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 03/29/2022] [Accepted: 04/25/2022] [Indexed: 02/07/2023] Open
Abstract
Lung transplantation can improve the survival of patients with severe chronic pulmonary disorders. However, the short- and long-term risk of infections can increase morbidity and mortality rates. A non-systematic review was performed to provide the most updated information on pathogen, host, and environment-related factors associated with the occurrence of bacterial, fungal, and viral infections as well as the most appropriate therapeutic options. Bacterial infections account for about 50% of all infectious diseases in lung transplanted patients, while viruses represent the second cause of infection accounting for one third of all infections. Almost 10% of patients develop invasive fungal infections during the first year after lung transplant. Pre-transplantation comorbidities, disruption of physical barriers during the surgery, and exposure to nosocomial pathogens during the hospital stay are directly associated with the occurrence of life-threatening infections. Empiric antimicrobial treatment after the assessment of individual risk factors, local epidemiology of drug-resistant pathogens and possible drug-drug interactions can improve the clinical outcomes.
Collapse
Affiliation(s)
- M Dettori
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - N Riccardi
- StopTB Italia Onlus, Milan, Italy; Department of Clinical and Experimental Medicine, University of Pisa, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - D Canetti
- StopTB Italia Onlus, Milan, Italy; Department of Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - R M Antonello
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, Trieste, Italy
| | - A F Piana
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - A Palmieri
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - P Castiglia
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - A A Azara
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - M D Masia
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - A Porcu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - G C Ginesu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - M L Cossu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - M Conti
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - P Pirina
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - A Fois
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - I Maida
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - G Madeddu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - S Babudieri
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - L Saderi
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - G Sotgiu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy; StopTB Italia Onlus, Milan, Italy.
| |
Collapse
|
11
|
Amjad W, Hamaad Rahman S, Schiano TD, Jafri SM. Epidemiology and Management of Infections in Liver Transplant Recipients. Surg Infect (Larchmt) 2024; 25:272-290. [PMID: 38700753 DOI: 10.1089/sur.2023.346] [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] [Indexed: 07/13/2024] Open
Abstract
Background: Improvements in liver transplant (LT) outcomes are attributed to advances in surgical techniques, use of potent immunosuppressants, and rigorous pre-LT testing. Despite these improvements, post-LT infections remain the most common complication in this population. Bacteria constitute the most common infectious agents, while fungal and viral infections are also frequently encountered. Multi-drug-resistant bacterial infections develop because of polymicrobial overuse and prolonged hospital stays. Immediate post-LT infections are commonly caused by viruses. Conclusions: Appropriate vaccination, screening of both donor and recipients before LT and antiviral prophylaxis in high-risk individuals are recommended. Antimicrobial drug resistance is common in high-risk LT and associated with poor outcomes; epidemiology and management of these cases is discussed. Additionally, we also discuss the effect of coronavirus disease 2019 (COVID-19) infection and monkeypox in the LT population.
Collapse
Affiliation(s)
- Waseem Amjad
- Gastroenterology and Hepatology, University of Maryland, Baltimore, Maryland, USA
| | | | - Thomas D Schiano
- Recanati-Miller Transplantation Institute, Division of Liver Diseases, Mount Sinai Medical Center, New York, New York, USA
| | - Syed-Mohammed Jafri
- Gastroenterology and Hepatology, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
12
|
Schultz BG, Kotton CN, Jutlla G, Ressa R, de Lacey T, Chowdhury E, Bo T, Fenu E, Gelone DK, Poirrier JE, Amorosi SL. Cost-effectiveness of maribavir versus conventional antiviral therapies for post-transplant refractory cytomegalovirus infection with or without genotypic resistance: A US perspective. J Med Virol 2024; 96:e29609. [PMID: 38647051 DOI: 10.1002/jmv.29609] [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: 12/12/2023] [Revised: 03/20/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
This study evaluated the cost-effectiveness of maribavir versus investigator-assigned therapy (IAT; valganciclovir/ganciclovir, foscarnet, or cidofovir) for post-transplant refractory cytomegalovirus (CMV) infection with or without resistance. A two-stage Markov model was designed using data from the SOLSTICE trial (NCT02931539), real-world multinational observational studies, and published literature. Stage 1 (0-78 weeks) comprised clinically significant CMV (csCMV), non-clinically significant CMV (n-csCMV), and dead states; stage 2 (78 weeks-lifetime) comprised alive and dead states. Total costs (2022 USD) and quality-adjusted life years (QALYs) were estimated for the maribavir and IAT cohorts. An incremental cost-effectiveness ratio was calculated to determine cost-effectiveness against a willingness-to-pay threshold of $100 000/QALY. Compared with IAT, maribavir had lower costs ($139 751 vs $147 949) and greater QALYs (6.04 vs 5.83), making it cost-saving and more cost-effective. Maribavir had higher acquisition costs compared with IAT ($80 531 vs $65 285), but lower costs associated with administration/monitoring ($16 493 vs $27 563), adverse events (AEs) ($11 055 vs $16 114), hospitalization ($27 157 vs $33 905), and graft loss ($4516 vs $5081), thus making treatment with maribavir cost-saving. Maribavir-treated patients spent more time without CMV compared with IAT-treated patients (0.85 years vs 0.68 years), leading to lower retreatment costs for maribavir (cost savings: -$42 970.80). Compared with IAT, maribavir was more cost-effective for transplant recipients with refractory CMV, owing to better clinical efficacy and avoidance of high costs associated with administration, monitoring, AEs, and hospitalizations. These results can inform healthcare decision-makers on the most effective use of their resources for post-transplant refractory CMV treatment.
Collapse
Affiliation(s)
- Bob G Schultz
- Takeda Pharmaceuticals U.S.A., Inc., Lexington, Massachusetts, USA
| | - Camille N Kotton
- Infectious Diseases Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ginita Jutlla
- Parexel, Health Economics and Outcomes Research Modeling, London, UK
| | - Riccardo Ressa
- Parexel, Health Economics and Outcomes Research Modeling, London, UK
| | - Tam de Lacey
- Parexel, Health Economics and Outcomes Research Modeling, London, UK
| | - Emtiyaz Chowdhury
- Parexel, Health Economics and Outcomes Research Modeling, London, UK
| | - Tien Bo
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| | | | - Daniele K Gelone
- Takeda Pharmaceuticals U.S.A., Inc., Lexington, Massachusetts, USA
| | | | - Stacey L Amorosi
- Takeda Development Center Americas, Inc., Lexington, Massachusetts, USA
| |
Collapse
|
13
|
Bottino P, Pastrone L, Zanotto E, Sidoti F, Cavallo R, Costa C. Molecular diagnosis of Cytomegalovirus infection: clinical performance of the Aptima transcription-mediated amplification assay toward conventional qPCR chemistry on whole blood samples. J Clin Microbiol 2024; 62:e0090623. [PMID: 38349144 PMCID: PMC10935658 DOI: 10.1128/jcm.00906-23] [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: 07/17/2023] [Accepted: 01/16/2024] [Indexed: 03/14/2024] Open
Abstract
Human Cytomegalovirus (HCMV) infection is life-threatening for immunocompromised patients. Quantitative molecular assays on whole blood or plasma are the gold standard for the diagnosis of invasive HCMV infection and for monitoring antiviral treatment in individuals at risk of HCMV disease. For these reasons, an accurate standardization toward the WHO 1st International Standard among different centers and diagnostic kits represents an effort for better clinical management of HCMV-positive patients. Herein, we evaluate, for the first time, the performance of a new transcription-mediated amplification (TMA) assay versus quantitative polymerase chain reaction (qPCR) chemistry, used as a routine method, on whole blood samples. A total of 755 clinical whole blood specimens were collected and tested simultaneously with TMA and qPCR assays. The data showed a qualitative agreement of 99.27% for positive quantified samples and 89.39% for those undetected between the two tested methods. Evaluation of viremia in positive samples highlighted a good correlation between TMA and qPCR chemistries in terms of International Units (ΔLog10 IU/mL: -0.29 ± 0.40). The TMA assay showed a significant correlation with qPCR in patients monitored for up to 3 months, thus allowing an accurate assessment of viremia in transplant patients. Therefore, TMA chemistry showed good agreement with qPCR testing, used as a current diagnostic routine. It also offers important advantages, such as FDA approval on plasma and In Vitro Diagnostic (IVD) on both plasma and whole blood, automated workflow with minimal hands-on time, and random access loading, thus enabling a rapid and reliable diagnostic in HCMV-infected patients. IMPORTANCE In this paper, we describe the clinical performance of a novel transcription-mediated amplification (TMA) assay for the detection and quantification of human Cytomegalovirus (HCMV) DNA from whole blood samples. This is a pivotal analysis in immunocompromised patients [transplanted, HIV-positive, and Hematopoietic Stem Cell (HSC) recipients], and molecular tests with high sensitivity and specificity are necessary to evaluate the HCMV viral load in these patients. To our knowledge, this is the first in-depth evaluation of TMA chemistry for HCMV diagnosis on whole blood samples. Moreover, also technical aspects of this assay make it suitable for clinical diagnostics.
Collapse
Affiliation(s)
- Paolo Bottino
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Lisa Pastrone
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Elisa Zanotto
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Francesca Sidoti
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Rossana Cavallo
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Cristina Costa
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| |
Collapse
|
14
|
Magda G. Opportunistic Infections Post-Lung Transplantation: Viral, Fungal, and Mycobacterial. Infect Dis Clin North Am 2024; 38:121-147. [PMID: 38280760 DOI: 10.1016/j.idc.2023.12.001] [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] [Indexed: 01/29/2024]
Abstract
Opportunistic infections are a leading cause of lung transplant recipient morbidity and mortality. Risk factors for infection include continuous exposure of the lung allograft to the external environment, high levels of immunosuppression, impaired mucociliary clearance and decreased cough reflex, and impact of the native lung microbiome in single lung transplant recipients. Infection risk is mitigated through careful pretransplant screening of recipients and donors, implementation of antimicrobial prophylaxis strategies, and routine surveillance posttransplant. This review describes common viral, fungal, and mycobacterial infectious after lung transplant and provides recommendations on prevention and treatment.
Collapse
Affiliation(s)
- Gabriela Magda
- Columbia University Lung Transplant Program, Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, Columbia University Vagelos College of Physicians and Surgeons, 622 West 168th Street PH-14, New York, NY 10032, USA.
| |
Collapse
|
15
|
Abidi MZ, Schold JD, Kaplan B, Weinberg A, Erlandson KM, Malamon JS. Patient years lost due to cytomegalovirus serostatus mismatching in the scientific registry of transplant recipients. Front Immunol 2024; 14:1292648. [PMID: 38264645 PMCID: PMC10803440 DOI: 10.3389/fimmu.2023.1292648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Background The cytomegalovirus (CMV) mismatch rate in deceased donor kidney transplant (DDKT) recipients in the US remains above 40%. Since CMV mismatching is common in DDKT recipients, the cumulative effects may be significant in the context of overall patient and graft survival. Our primary objective was to describe the short- and long-term risks associated with high-risk CMV donor positive/recipient negative (D+/R-) mismatching among DDKT recipients with the explicit goal of deriving a mathematical mismatching penalty. Methods We conducted a retrospective, secondary analysis of the Scientific Registry of Transplant Recipients (SRTR) database using donor-matched DDKT recipient pairs (N=105,608) transplanted between 2011-2022. All-cause mortality and graft failure hazard ratios were calculated from one year to ten years post-DDKT. All-cause graft failure included death events. Survival curves were calculated using the Kaplan-Meier estimation at 10 years post-DDKT and extrapolated to 20 years to provide the average graft days lost (aGDL) and average patient days lost (aPDL) due to CMV D+/R- serostatus mismatching. We also performed an age-based stratification analysis to compare the relative risk of CMV D+ mismatching by age. Results Among 31,518 CMV D+/R- recipients, at 1 year post-DDKT, the relative risk of death increased by 29% (p<0.001), and graft failure increased by 17% (p<0.001) as compared to matched CMV D+/R+ group (N=31,518). Age stratification demonstrated a significant increase in the risk associated with CMV mismatching in patients 40 years of age and greater. The aGDL per patient due to mismatching was 125 days and the aPDL per patient was 100 days. Conclusion The risks of CMV D+/R- mismatching are seen both at 1 year post-DDKT period and accumulated throughout the lifespan of the patient, with the average CMV D+/R- recipient losing more than three months of post-DDKT survival time. CMV D+/R- mismatching poses a more significant risk and a greater health burden than previously reported, thus obviating the need for better preventive strategies including CMV serodirected organ allocation to prolong lifespans and graft survival in high-risk patients.
Collapse
Affiliation(s)
- Maheen Z. Abidi
- Department of Medicine, Division of Infectious Diseases, University of Colorado, Aurora, CO, United States
| | - Jesse D. Schold
- Department of Surgery, Division of Transplant Surgery, University of Colorado, Aurora, CO, United States
- Colorado Center for Transplantation Care (CCTCARE), Research and Education, Division of Transplant Surgery, Department of Surgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Bruce Kaplan
- Department of Surgery, Division of Transplant Surgery, University of Colorado, Aurora, CO, United States
- Colorado Center for Transplantation Care (CCTCARE), Research and Education, Division of Transplant Surgery, Department of Surgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Adriana Weinberg
- Department of Medicine, Division of Infectious Diseases, University of Colorado, Aurora, CO, United States
- Department of Pediatrics and Pathology, University of Colorado, Aurora, CO, United States
| | - Kristine M. Erlandson
- Department of Medicine, Division of Infectious Diseases, University of Colorado, Aurora, CO, United States
| | - John S. Malamon
- Department of Surgery, Division of Transplant Surgery, University of Colorado, Aurora, CO, United States
- Colorado Center for Transplantation Care (CCTCARE), Research and Education, Division of Transplant Surgery, Department of Surgery, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| |
Collapse
|
16
|
Fayyaz A, Raja M, Natori Y. Prevention and Management of Infections in Lung Transplant Recipients. J Clin Med 2023; 13:11. [PMID: 38202018 PMCID: PMC10779253 DOI: 10.3390/jcm13010011] [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: 09/24/2023] [Revised: 11/13/2023] [Accepted: 12/01/2023] [Indexed: 01/12/2024] Open
Abstract
Anti-rejection medications are essential in preventing organ rejection amongst solid organ transplant recipients; however, these agents also cause profound immunosuppression, predisposing lung transplant recipients (LTRs) to infectious complications. The timely management including prevention, diagnosis, and treatment of such infectious complications is vital to prevent significant morbidity and mortality in solid organ transplant recipients and allograft dysfunction. LTRs are inundated with microbes that may be recognized as commensals in hosts with intact immune systems. Bacterial infections are the most common ones, followed by viral pathogens. Indications of a brewing infectious process may be subtle. Hence, the importance of adapting vigilance around isolated hints through symptomatology and signs is pivotal. Signals to suggest an infectious process, such as fever and leukocytosis, may be dampened by immunosuppressive agents. One must also be vigilant about drug interactions of antibiotics and immunosuppressive agents. Treatment of infections can become challenging, as antimicrobials can interact with immunosuppressive agents, and antimicrobial resistance can surge under antimicrobial pressure. Transplant infectious disease physicians work in concert with transplant teams to obtain specimens for diagnostic testing and follow through with source control when possible. This heavily impacts medical decisions and fosters a multidisciplinary approach in management. Furthermore, the reduction of immunosuppression, although it augments the risk of allograft rejection, is as crucial as the initiation of appropriate antimicrobials when it comes to the management of infections.
Collapse
Affiliation(s)
| | | | - Yoichiro Natori
- Miami Transplant Institute, Jackson Health System, Division of Infectious Disease, Department of Clinical Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.F.); (M.R.)
| |
Collapse
|
17
|
Elalouf A, Elalouf H, Rosenfeld A. Modulatory immune responses in fungal infection associated with organ transplant - advancements, management, and challenges. Front Immunol 2023; 14:1292625. [PMID: 38143753 PMCID: PMC10748506 DOI: 10.3389/fimmu.2023.1292625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Organ transplantation stands as a pivotal achievement in modern medicine, offering hope to individuals with end-stage organ diseases. Advancements in immunology led to improved organ transplant survival through the development of immunosuppressants, but this heightened susceptibility to fungal infections with nonspecific symptoms in recipients. This review aims to establish an intricate balance between immune responses and fungal infections in organ transplant recipients. It explores the fundamental immune mechanisms, recent advances in immune response dynamics, and strategies for immune modulation, encompassing responses to fungal infections, immunomodulatory approaches, diagnostics, treatment challenges, and management. Early diagnosis of fungal infections in transplant patients is emphasized with the understanding that innate immune responses could potentially reduce immunosuppression and promise efficient and safe immuno-modulating treatments. Advances in fungal research and genetic influences on immune-fungal interactions are underscored, as well as the potential of single-cell technologies integrated with machine learning for biomarker discovery. This review provides a snapshot of the complex interplay between immune responses and fungal infections in organ transplantation and underscores key research directions.
Collapse
Affiliation(s)
- Amir Elalouf
- Department of Management, Bar-Ilan University, Ramat Gan, Israel
| | - Hadas Elalouf
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| | - Ariel Rosenfeld
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
18
|
Bottino P, Pastrone L, Curtoni A, Bondi A, Sidoti F, Zanotto E, Cavallo R, Solidoro P, Costa C. Antiviral Approach to Cytomegalovirus Infection: An Overview of Conventional and Novel Strategies. Microorganisms 2023; 11:2372. [PMID: 37894030 PMCID: PMC10608897 DOI: 10.3390/microorganisms11102372] [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: 08/28/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a herpesvirus capable of establishing a lifelong persistence in the host through a chronic state of infection and remains an essential global concern due to its distinct life cycle, mutations, and latency. It represents a life-threatening pathogen for immunocompromised patients, such as solid organ transplanted patients, HIV-positive individuals, and hematopoietic stem cell recipients. Multiple antiviral approaches are currently available and administered in order to prevent or manage viral infections in the early stages. However, limitations due to side effects and the onset of antidrug resistance are a hurdle to their efficacy, especially for long-term therapies. Novel antiviral molecules, together with innovative approaches (e.g., genetic editing and RNA interference) are currently in study, with promising results performed in vitro and in vivo. Since HCMV is a virus able to establish latent infection, with a consequential risk of reactivation, infection management could benefit from preventive treatment for critical patients, such as immunocompromised individuals and seronegative pregnant women. This review will provide an overview of conventional antiviral clinical approaches and their mechanisms of action. Additionally, an overview of proposed and developing new molecules is provided, including nucleic-acid-based therapies and immune-mediated approaches.
Collapse
Affiliation(s)
- Paolo Bottino
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Lisa Pastrone
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Antonio Curtoni
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Alessandro Bondi
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Francesca Sidoti
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Elisa Zanotto
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Rossana Cavallo
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Paolo Solidoro
- Pneumology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy;
| | - Cristina Costa
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| |
Collapse
|
19
|
Mafi S, Alain S, Hantz S. Evaluation of the fully automated LIAISON®XL chemiluminescence analyzer for QuantiFERON®-CMV testing in transplant recipients. J Clin Virol 2023; 166:105550. [PMID: 37527584 DOI: 10.1016/j.jcv.2023.105550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Monitoring CMV-specific cell-mediated immunity by the QuantiFERON®-CMV (QF-CMV) may be useful in predicting the risk of CMV infection in transplant recipients (TR). OBJECTIVES As the QuantiFERON-Tuberculosis (QFT®-Plus) became available on the fully automated LIAISON®XL chemiluminescence (CLIA) analyzer, we evaluated the performance of the QF-CMV on the LIAISON®XL analyzer using the QuantiFERON®-TB Gold Plus reagent. STUDY DESIGN Between 2018 and 2022, 81 samples from TR were collected at the Department of Virology of Limoges Hospital, France. Whole blood was collected into each of the three QF-CMV collection tubes: a CMV-antigen tube (QF-Ag), a mitogen tube (QF-Mg) (positive control), and a nil tube (negative control). The QF-CMV was performed on the LIAISON®XL analyzer, and results were compared with those obtained by conventional microplate ELISA. RESULTS Intra- and inter-assay coefficients of variation were inferior to 20%. No inter-sample contamination was found (p=0.366). The level of concordance between CLIA and the commonly used ELISA method was 88.9%. Positive and negative percent agreements were 92.3% and 85.7%, respectively, with a very good agreement between assays (κ=0.818). Most discordances were due to indeterminate- or negative-ELISA/positive-CLIA results (most of ELISA results were borderline). Linear regression analyses demonstrated a strong correlation between both assays (QF-Ag Pearson's r=0.978, QF-Mg Pearson's r=0.963). No significant difference was observed in median QF-CMV values between both assays (QF-Ag p=0.776; QF-Mg p=0.853; Mann-Whitney U test). The Bland-Altman plots showed a minor difference in IFN-γ release (QF-Ag -0.069 IU/ml, 95% limits of agreement (LoA): -1.589; 1.451; QF-Mg 0.190 IU/ml, 95% LoA: -2.070; 2.450). CONCLUSION Automated QF-CMV with CLIA is comparable to QF-CMV performed by ELISA with a presumably higher sensitivity for IFN-γ detection that may result in the conversion of samples close to the ELISA cut-off into positive results. Moreover, the use of a random-access analyzer allows to optimize the follow-up of TR.
Collapse
Affiliation(s)
- Sarah Mafi
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, CHU Limoges, F-87000 Limoges, France; INSERM, RESINFIT, U1092, F-87000, Limoges, France.
| | - Sophie Alain
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, CHU Limoges, F-87000 Limoges, France; INSERM, RESINFIT, U1092, F-87000, Limoges, France
| | - Sébastien Hantz
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, CHU Limoges, F-87000 Limoges, France; INSERM, RESINFIT, U1092, F-87000, Limoges, France.
| |
Collapse
|
20
|
Breitkopf R, Treml B, Bukumiric Z, Innerhofer N, Fodor M, Radovanovic Spurnic A, Rajsic S. Cytomegalovirus Disease as a Risk Factor for Invasive Fungal Infections in Liver Transplant Recipients under Targeted Antiviral and Antimycotic Prophylaxis. J Clin Med 2023; 12:5198. [PMID: 37629240 PMCID: PMC10455861 DOI: 10.3390/jcm12165198] [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: 06/27/2023] [Revised: 07/27/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Cytomegalovirus (CMV) infection is the most common opportunistic infection that occurs following orthotopic liver transplantation (OLT). In addition to the direct infection-related symptoms, it also triggers an immunological response that may contribute to adverse clinical outcomes. CMV disease has been described as a predictor of invasive fungal infections (IFIs) but its role under an antiviral prophylaxis regimen is unclear. METHODS We retrospectively analyzed the medical records of 214 adult liver transplant recipients (LTRs). Universal antiviral prophylaxis was utilized in recipients with CMV mismatch; intermediate- and low-risk patients received pre-emptive treatment. RESULTS Six percent of patients developed CMV disease independent of their serostatus. The occurrence of CMV disease was associated with elevated virus load and increased incidence of leucopenia and IFIs. Furthermore, CMV disease was associated with higher one-year mortality and increased relapse rates within the first year of OLT. CONCLUSIONS CMV disease causes significant morbidity and mortality in LTRs, directly affecting transplant outcomes. Due to the increased risk of IFIs, antifungal prophylaxis for CMV disease may be appropriate. Postoperative CMV monitoring should be considered after massive transfusion, even in low-risk serostatus constellations. In case of biliary complications, biliary CMV monitoring may be appropriate in the case of CMV-DNA blood-negative patients.
Collapse
Affiliation(s)
- Robert Breitkopf
- Department of Anesthesia and Intensive Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria (N.I.)
| | - Benedikt Treml
- Department of Anesthesia and Intensive Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria (N.I.)
| | - Zoran Bukumiric
- Institute of Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nicole Innerhofer
- Department of Anesthesia and Intensive Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria (N.I.)
| | - Margot Fodor
- Department of Visceral, Transplantation and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | | | - Sasa Rajsic
- Department of Anesthesia and Intensive Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria (N.I.)
| |
Collapse
|
21
|
Lambert N, El Moussaoui M, Baron F, Maquet P, Darcis G. Virus-Specific T-Cell Therapy for Viral Infections of the Central Nervous System: A Review. Viruses 2023; 15:1510. [PMID: 37515196 PMCID: PMC10383098 DOI: 10.3390/v15071510] [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: 06/08/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Opportunistic viral infections of the central nervous system represent a significant cause of morbidity and mortality among an increasing number of immunocompromised patients. Since antiviral treatments are usually poorly effective, the prognosis generally relies on the ability to achieve timely immune reconstitution. Hence, strategies aimed at reinvigorating antiviral immune activity have recently emerged. Among these, virus-specific T-cells are increasingly perceived as a principled and valuable tool to treat opportunistic viral infections. Here we briefly discuss how to develop and select virus-specific T-cells, then review their main indications in central nervous system infections, including progressive multifocal leukoencephalopathy, CMV infection, and adenovirus infection. We also discuss their potential interest in the treatment of progressive multiple sclerosis, or EBV-associated central nervous system inflammatory disease. We finish with the key future milestones of this promising treatment strategy.
Collapse
Affiliation(s)
- Nicolas Lambert
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Majdouline El Moussaoui
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| | - Frédéric Baron
- Department of Hematology, University Hospital of Liège, 4000 Liège, Belgium
| | - Pierre Maquet
- Department of Neurology, University Hospital of Liège, 4000 Liège, Belgium
| | - Gilles Darcis
- Department of General Internal Medicine and Infectious Diseases, University Hospital of Liège, 4000 Liège, Belgium
| |
Collapse
|
22
|
Møller DL, Sørensen SS, Rezahosseini O, Rasmussen DB, Arentoft NS, Loft JA, Perch M, Gustafsson F, Lundgren J, Scheike T, Knudsen JD, Ostrowski SR, Rasmussen A, Nielsen SD. Prediction of herpes virus infections after solid organ transplantation: a prospective study of immune function. Front Immunol 2023; 14:1183703. [PMID: 37465673 PMCID: PMC10351284 DOI: 10.3389/fimmu.2023.1183703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/24/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Herpes virus infections are a major concern after solid organ transplantation and linked to the immune function of the recipient. We aimed to determine the incidence of positive herpes virus (cytomegalovirus (CMV), Epstein-Barr virus (EBV), herpes simplex virus type 1/2 (HSV-1/2), and varicella zoster virus (VZV)) PCR tests during the first year post-transplantation and assess whether a model including immune function pre-transplantation and three months post-transplantation could predict a subsequent positive herpes virus PCR. Methods All participants were preemptively screened for CMV, and EBV IgG-negative participants were screened for EBV during the first year post-transplantation. Herpes virus PCR tests for all included herpes viruses (CMV, EBV, HSV-1/2, and VZV) were retrieved from the Danish Microbiology database containing nationwide PCR results from both hospitals and outpatient clinics. Immune function was assessed by whole blood stimulation with A) LPS, B) R848, C) Poly I:C, and D) a blank control. Cytokine concentrations (TNF-α, IL-1β, IL-6, IL-8, IL-10, IL-12p40, IL-17A, IFN-α, and IFN-γ) were measured using Luminex. Results We included 123 liver (54%), kidney (26%), and lung (20%) transplant recipients. The cumulative incidence of positive herpes virus PCR tests was 36.6% (95% CI: 28.1-45.1) during the first year post-transplantation. The final prediction model included recipient age, type of transplantation, CMV serostatus, and change in Poly I:C-induced IL-12p40 from pre-transplantation to three months post-transplantation. The prediction model had an AUC of 77% (95% CI: 61-92). Risk scores were extracted from the prediction model, and the participants were divided into three risk groups. Participants with a risk score <5 (28% of the cohort), 5-10 (45% of the cohort), and >10 (27% of the cohort) had a cumulative incidence of having a positive herpes virus PCR test at 5.8%, 25%, and 73%, respectively (p < 0.001). Conclusion In conclusion, the incidence of positive herpes virus PCR tests was high, and a risk model including immune function allowed the prediction of positive herpes virus PCR and may be used to identify recipients at higher risk.
Collapse
Affiliation(s)
- Dina Leth Møller
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Søren Schwartz Sørensen
- Department of Nephrology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Omid Rezahosseini
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Bräuner Rasmussen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nicoline Stender Arentoft
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Josefine Amalie Loft
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Michael Perch
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Section for Lung Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Finn Gustafsson
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lundgren
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Centre of Excellence for Health, Immunity, and Infections, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Scheike
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Jenny Dahl Knudsen
- Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Allan Rasmussen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Viro-immunology Research Unit, Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Mafi S, Essig M, Rerolle JP, Lagathu G, Crochette R, Brodard V, Schvartz B, Gouarin S, Bouvier N, Engelmann I, Garstka A, Bressollette-Bodin C, Cantarovitch D, Germi R, Janbon B, Archimbaut C, Heng AE, Garnier F, Gomes-Mayeras M, Labrunie A, Hantz S, Alain S. Torque teno virus viremia and QuantiFERON ®-CMV assay in prediction of cytomegalovirus reactivation in R+ kidney transplant recipients. Front Med (Lausanne) 2023; 10:1180769. [PMID: 37425298 PMCID: PMC10323437 DOI: 10.3389/fmed.2023.1180769] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/12/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Cytomegalovirus (CMV) is the most frequent infectious complication following solid organ transplantation. Torque teno viruses (TTV) viremia has been proposed as a biomarker of functional immunity in the management of kidney transplant recipients (KTR). The QuantiFERON®-CMV (QF-CMV) is a commercially available assay that allows the assessment of CD8+ T-cell responses in routine diagnostic laboratories. Methods In a prospective national multicenter cohort of 64 CMV-seropositive (R+) KTR, we analyzed the value of TTV load and the two markers of the QF-CMV assay [QF-Ag (CMV-specific T-cell responses) and QF-Mg (overall T-cell responses)], alone and in combination, in prediction of CMV reactivation (≥3 log10 IU/ ml) in the first post-transplant year. We compared previously published cut-offs and specific cut-offs optimized from ROC curves for our population. Results Using the conventional cut-off (3.45 log10 copies/ml), TTV load at D0 [inclusion visit on the day of transplantation before induction (D0)], or at M1 (1-month post-transplant visit) perform better in predicting CMV viremia control than CMV reactivation. Survival analyses suggest a better performance of our optimized TTV cut-offs (3.78 log10 copies/ml at D0 and 4.23 log10 copies/ml at M1) for risk stratification of CMV reactivation in our R+ KTR cohort. The QF-CMV (QF-Ag = 0.2 IU/ml, and QF-Mg = 0.5 IU/ml) also appears to better predict CMV viremia control than CMV reactivation. Moreover, survival analyses suggest that the QF-Mg would perform better than the QF-Ag in stratifying the risk of CMV reactivation. The use of our optimized QF-Mg cut-off (1.27 IU/ml) at M1 further improved risk stratification of CMV reactivation. Using conventional cut-offs, the combination of TTV load and QF-Ag or TTV load and QF-Mg did not improve prediction of CMV viremia control compared to separate analysis of each marker but resulted in an increase of positive predictive values. The use of our cut-offs slightly improved risk prediction of CMV reactivation. Conclusion The combination of TTV load and QF-Ag or TTV load and QF-Mg could be useful in stratifying the risk of CMV reactivation in R+ KTR during the first post-transplant year and thereby have an impact on the duration of prophylaxis in these patients. Clinical trial registration ClinicalTrials.gov registry, identifier NCT02064699.
Collapse
Affiliation(s)
- Sarah Mafi
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
- Inserm, RESINFIT, U1092, Université de Limoges, Limoges, France
| | - Marie Essig
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Jean-Philippe Rerolle
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Gisèle Lagathu
- Virology Department, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Romain Crochette
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Véronique Brodard
- Virology Department, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Betoul Schvartz
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Reims, Reims, France
| | - Stephanie Gouarin
- Virology Department, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Nicolas Bouvier
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Ilka Engelmann
- Virology Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Antoine Garstka
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Lille, Lille, France
| | | | - Diego Cantarovitch
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Raphaële Germi
- Virology Department, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Benedicte Janbon
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Grenoble, Grenoble, France
| | - Christine Archimbaut
- Virology Department, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Anne-Elizabeth Heng
- Nephrology and Transplantation Department, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Françoise Garnier
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
- Inserm, RESINFIT, U1092, Université de Limoges, Limoges, France
| | - Melissa Gomes-Mayeras
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
- Inserm, RESINFIT, U1092, Université de Limoges, Limoges, France
| | - Anaïs Labrunie
- Biostatistics Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
| | - Sébastien Hantz
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
- Inserm, RESINFIT, U1092, Université de Limoges, Limoges, France
| | - Sophie Alain
- French National Reference Center for Herpesviruses, Bacteriology, Virology, Hygiene Department, Centre Hospitalier Universitaire de Limoges, Limoges, France
- Inserm, RESINFIT, U1092, Université de Limoges, Limoges, France
| |
Collapse
|
24
|
Ribeiro RV, Samman A, Wang A, Wang S, Martinu T, Keshavjee S, Singer LG, Kumar D, Humar A, Cypel M. Incidence of post-transplant cytomegalovirus viremia in patients receiving lungs after ex vivo lung perfusion. JTCVS OPEN 2023; 14:590-601. [PMID: 37425481 PMCID: PMC10328819 DOI: 10.1016/j.xjon.2023.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 07/11/2023]
Abstract
Objectives Cytomegalovirus infection after lung transplant is associated with increased morbidity and mortality. Inflammation, infection, and longer ischemic times are important risk factors for cytomegalovirus infection. Ex vivo lung perfusion has helped to successfully increase the use of high-risk donors over the last decade. However, the impact of ex vivo lung perfusion on post-transplant cytomegalovirus infection is unknown. Methods We performed a retrospective analysis of all adult lung transplant recipients from 2010 to 2020. The primary end point was comparison of cytomegalovirus viremia between patients who received ex vivo lung perfusion donor lungs and patients who received non-ex vivo lung perfusion donor lungs. Cytomegalovirus viremia was defined as cytomegalovirus viral load greater than 1000 IU/mL within 2 years post-transplant. Secondary end points were the time from lung transplant to cytomegalovirus viremia, peak cytomegalovirus viral load, and survival. Outcomes were also compared between the different donor recipient cytomegalovirus serostatus matching groups. Results Included were 902 recipients of non-ex vivo lung perfusion lungs and 403 recipients of ex vivo lung perfusion lungs. There was no significant difference in the distribution of the cytomegalovirus serostatus matching groups. A total of 34.6% of patients in the non-ex vivo lung perfusion group developed cytomegalovirus viremia, as did 30.8% in the ex vivo lung perfusion group (P = .17). There was no difference in time to viremia, peak viral loads, or survival when comparing both groups. Likewise, all outcomes were comparable in the non-ex vivo lung perfusion and ex vivo lung perfusion groups within each serostatus matching group. Conclusions The practice of using more injured donor organs via ex vivo lung perfusion has not affected cytomegalovirus viremia rates and severity in lung transplant recipients in our center.
Collapse
Affiliation(s)
- Rafaela V.P. Ribeiro
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Anas Samman
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Aizhou Wang
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Stella Wang
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Lianne G. Singer
- Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Deepali Kumar
- Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Atul Humar
- Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Lalagkas PN, Iliou J, Rigo R, Miarons M, Fernández-Alarcon B, Bestard O, Cruzado JM, Melilli E, Torras J, Grinyó JM, Lloberas N, Colom H. Comparison of Three Renal Function Formulas for Ganciclovir/Valganciclovir Dose Individualization in CMV-Infected Solid Organ Transplantation Patients Using a Population Approach. Clin Pharmacokinet 2023; 62:861-880. [PMID: 37140726 PMCID: PMC10256675 DOI: 10.1007/s40262-023-01237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND AND OBJECTIVE The gold standard treatment of established cytomegalovirus infection or prevention in solid organ transplantation is the intravenous administration of ganciclovir (GCV) or oral administration of valganciclovir (VGCV), both adjusted to the renal function. In both instances, there is a high interindividual pharmacokinetic variability, mainly owing to the wide range of variation of both the renal function and body weight. Therefore, accurate estimation of the renal function is crucial for GCV/VGCV dose optimization. This study aimed to compare three different formulas for estimating the renal function in solid organ transplantation patients with cytomegalovirus infection, for individualizing antiviral therapy with GCV/VGCV, using a population approach. METHODS A population pharmacokinetic analysis was performed using NONMEM 7.4. A total of 650 plasma concentrations obtained after intravenous GCV and oral VGCV administrations were analyzed, from intensive and sparse sampling designs. Three different population pharmacokinetic models were built with the renal function given by Cockcroft-Gault, Modification of Diet in Renal Disease, or Chronic Kidney Disease EPIdemiology Collaboration (CKD-EPI) formulas. Pharmacokinetic parameters were allometrically scaled to body weight. RESULTS The CKD-EPI formula was identified as the best predictor of between-patient variability in GCV clearance. Internal and external validation techniques showed that the CKD-EPI model had better stability and performed better compared with the others. CONCLUSIONS The model based on the more accurate estimation of the renal function with the CKD-EPI formula and body weight as a size metric most used in the clinical practice can refine initial dose recommendations and contribute to GCV and VGCV dose individualization when required in the prevention or treatment of cytomegalovirus infection in solid organ transplantation patients.
Collapse
Affiliation(s)
- Panagiotis Nikolaos Lalagkas
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain
| | - Jorge Iliou
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain
| | - Raul Rigo
- Biochemistry Department, IDIBELL, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Marta Miarons
- Pharmacy Service, Hospital de la Vall d'Hebron, Barcelona, Spain
| | - Beatriz Fernández-Alarcon
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain
| | - Oriol Bestard
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Josep M Cruzado
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Edoardo Melilli
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Joan Torras
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Josep M Grinyó
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Nuria Lloberas
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain.
| | - Helena Colom
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain.
| |
Collapse
|
26
|
Sigdel TK, Boada P, Kerwin M, Rashmi P, Gjertson D, Rossetti M, Sur S, Munar D, Cimino J, Ahn R, Pickering H, Sen S, Parmar R, Fatou B, Steen H, Schaenman J, Bunnapradist S, Reed EF, Sarwal MM. Plasma proteome perturbation for CMV DNAemia in kidney transplantation. PLoS One 2023; 18:e0285870. [PMID: 37205661 PMCID: PMC10198483 DOI: 10.1371/journal.pone.0285870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 05/03/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) infection, either de novo or as reactivation after allotransplantation and chronic immunosuppression, is recognized to cause detrimental alloimmune effects, inclusive of higher susceptibility to graft rejection and substantive impact on chronic graft injury and reduced transplant survival. To obtain further insights into the evolution and pathogenesis of CMV infection in an immunocompromised host we evaluated changes in the circulating host proteome serially, before and after transplantation, and during and after CMV DNA replication (DNAemia), as measured by quantitative polymerase chain reaction (QPCR). METHODS LC-MS-based proteomics was conducted on 168 serially banked plasma samples, from 62 propensity score-matched kidney transplant recipients. Patients were stratified by CMV replication status into 31 with CMV DNAemia and 31 without CMV DNAemia. Patients had blood samples drawn at protocol times of 3- and 12-months post-transplant. Additionally, blood samples were also drawn before and 1 week and 1 month after detection of CMV DNAemia. Plasma proteins were analyzed using an LCMS 8060 triple quadrupole mass spectrometer. Further, public transcriptomic data on time matched PBMCs samples from the same patients was utilized to evaluate integrative pathways. Data analysis was conducted using R and Limma. RESULTS Samples were segregated based on their proteomic profiles with respect to their CMV Dnaemia status. A subset of 17 plasma proteins was observed to predict the onset of CMV at 3 months post-transplant enriching platelet degranulation (FDR, 4.83E-06), acute inflammatory response (FDR, 0.0018), blood coagulation (FDR, 0.0018) pathways. An increase in many immune complex proteins were observed at CMV infection. Prior to DNAemia the plasma proteome showed changes in the anti-inflammatory adipokine vaspin (SERPINA12), copper binding protein ceruloplasmin (CP), complement activation (FDR = 0.03), and proteins enriched in the humoral (FDR = 0.01) and innate immune responses (FDR = 0.01). CONCLUSION Plasma proteomic and transcriptional perturbations impacting humoral and innate immune pathways are observed during CMV infection and provide biomarkers for CMV disease prediction and resolution. Further studies to understand the clinical impact of these pathways can help in the formulation of different types and duration of anti-viral therapies for the management of CMV infection in the immunocompromised host.
Collapse
Affiliation(s)
- Tara K. Sigdel
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Patrick Boada
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Maggie Kerwin
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Priyanka Rashmi
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - David Gjertson
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Swastika Sur
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Dane Munar
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - James Cimino
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | - Richard Ahn
- Department of Microbiology and Immunology, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Subha Sen
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Benoit Fatou
- Department of Pathology, Harvard Medical School, Boston, MA, United States of America
| | - Hanno Steen
- Department of Pathology, Harvard Medical School, Boston, MA, United States of America
| | - Joanna Schaenman
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Suphamai Bunnapradist
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA, United States of America
| | - Minnie M. Sarwal
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States of America
| | | |
Collapse
|
27
|
Facchin A, Benyoub N, Elie V, Magreault S, Jacqz-Aigrain E. Limited Sampling Strategies to Predict Ganciclovir Exposure after Valganciclovir Administration and to Reduce Monitoring Constraints in Renal Transplant Children. Antimicrob Agents Chemother 2023; 67:e0159722. [PMID: 36880779 PMCID: PMC10112176 DOI: 10.1128/aac.01597-22] [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: 11/30/2022] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
Valganciclovir, the ganciclovir prodrug, is an antiviral agent used to prevent cytomegalovirus infection in renal transplant children. Therapeutic drug monitoring is still necessary to ensure optimal therapeutic area under the concentration-time curve from 0 to 24 h (AUC0-24) of 40 to 60 μg·h/mL since valganciclovir presents a high pharmacokinetic variability. To calculate ganciclovir AUC0-24 with the trapezoidal method, 7 samples are needed. The objective of this study was to develop and validate a reliable and clinically applicable limited sampling strategy (LSS) for individualizing valganciclovir dose in renal transplant children. Rich pharmacokinetic data from ganciclovir plasmatic dosages measured in renal transplant children who received valganciclovir to prevent cytomegalovirus infection at Robert Debré University Hospital were collected retrospectively. Ganciclovir AUC0-24s were calculated using the trapezoidal method. The LSS was developed using a multilinear regression approach to predict AUC0-24. The patients included were divided into two groups for model development (50 patients) and validation (30 patients). A total of 80 patients were included between February 2005 and November 2018. Multilinear regression models were developed on 50 pharmacokinetic profiles (50 patients) and validated with an independent group of 43 pharmacokinetic profiles (30 patients). Regressions based on samples collected at T1h-T4h-T8h, T2h-T4h-T8h, or T1h-T2h-T8h presented the best AUC0-24 predictive performances with an average difference between reference and predicted AUC0-24 of -0.27, 0.34, and -0.40 μg·h/mL, respectively. In conclusion, valganciclovir dosage adaptation was required in children to achieve the target AUC0-24. Three LSS models using three pharmacokinetic blood samples instead of seven will be useful for individualizing valganciclovir prophylaxis in renal transplant children.
Collapse
Affiliation(s)
- A. Facchin
- Department of Paediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, Assistance Publique – Hôpitaux de Paris, Paris, France
- Service of Pharmacy, Centre Hospitalier Intercommunal Nord Ardennes, Charleville-Mézières, France
| | - N. Benyoub
- Department of Paediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - V. Elie
- Department of Paediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - S. Magreault
- Department of Paediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - E. Jacqz-Aigrain
- Department of Paediatric Pharmacology and Pharmacogenetics, Hôpital Robert Debré, Assistance Publique – Hôpitaux de Paris, Paris, France
- University Paris -Cité, Paris, France
| |
Collapse
|
28
|
Opportunistic Infections Post-Lung Transplantation: Viral, Fungal, and Mycobacterial. Clin Chest Med 2023; 44:159-177. [PMID: 36774162 DOI: 10.1016/j.ccm.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Opportunistic infections are a leading cause of lung transplant recipient morbidity and mortality. Risk factors for infection include continuous exposure of the lung allograft to the external environment, high levels of immunosuppression, impaired mucociliary clearance and decreased cough reflex, and impact of the native lung microbiome in single lung transplant recipients. Infection risk is mitigated through careful pretransplant screening of recipients and donors, implementation of antimicrobial prophylaxis strategies, and routine surveillance posttransplant. This review describes common viral, fungal, and mycobacterial infectious after lung transplant and provides recommendations on prevention and treatment.
Collapse
|
29
|
Fifteen-Year Surveillance of LTR Receiving Pre-Emptive Therapy for CMV Infection: Prevention of CMV Disease and Incidence of CLAD. Microorganisms 2022; 10:microorganisms10122339. [PMID: 36557592 PMCID: PMC9788487 DOI: 10.3390/microorganisms10122339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
The efficacy of pre-emptive therapy in the prevention of cytomegalovirus (CMV) disease and the potential association of CMV infection with the occurrence of chronic lung allograft dysfunction (CLAD) was evaluated in 129 lung transplant recipients receiving pre-emptive therapy based on pp65-antigenemia or CMV-DNA in the blood and in the bronchoalveolar lavage. Seventy-one (55%) patients received pre-emptive ganciclovir/valganciclovir (GCV/VGCV) for CMV infection for a median of 28 (9-191) days. Possible CMV disease occurred in six (5%) patients and was healed after the GCV/VGCV therapy. The cumulative incidence of CLAD was 38% and 54% at 5 and 10 years. Acute rejection and CMV load in the blood (but not in the lung) were independent predictors of the occurrence of CLAD. Pre-emptive therapy is highly effective in preventing CMV disease in lung recipients and does not induce a superior incidence of CLAD compared to what reported for other cohorts of patients who received an extended antiviral prophylaxis.
Collapse
|
30
|
Montero C, Yomayusa N, Torres R, Cortes J, Alvarez C, Gallo J, Aldana G, Acevedo A, Rios M, Echeverri J, Yepes Z, Silva A, Gayon D, Perez J, Ibanez M. Low dose thymoglobulin versus basiliximab in cytomegalovirus positive kidney transplant recipients: Effectiveness of preemptive cytomegalovirus modified strategy. Nefrologia 2022:S2013-2514(22)00143-2. [PMID: 36437203 DOI: 10.1016/j.nefroe.2022.11.018] [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: 02/18/2021] [Accepted: 10/06/2021] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND We performed a retrospective trial to determine asymptomatic CMV reactivation and CMV disease in kidney allograft recipients with positive CMV serostatus. METHODS Preemptive modified strategy under low dose thymoglobulin versus basiliximab induction was evaluated. Patients were monitored by CMV-polymerase chain reaction (PCR); if the viral load was >4000copies/μl, they received valganciclovir adjusted for their renal function. RESULTS 132 recipients were included in the study, 84 and 48 receiving basiliximab and thymoglobulin induction respectively, and followed up for 12 months. Asymptomatic CMV reactivation was significantly higher for thymoglobulin (77.1% vs. 16.7%, p<0.001). Treatment groups had similar rates of CMV disease (3.6% vs. 2.1%, p 0.538). The significant difference in asymptomatic CMV reactivation between two treatment groups did not have any impact on 1 year graft function (71±26ml/min vs. 74±19ml/min; p=0.475) and no histological differences in protocol biopsies were observed among patients with asymptomatic CMV reactivation vs those without CMV reactivation. CONCLUSIONS Due to the high asymptomatic CMV reactivation incidence in patients who received thymoglobulin induction, our results suggest that valganciclovir prophylaxis may be advantageous in CMV seropositive renal transplant recipients after low dose thymoglobulin induction. A preemptive strategy appeared to significantly reduce the likelihood of CMV disease in both groups. Rejection risk and negative impact in renal function associated with asymptomatic CMV reactivation was not found in our series.
Collapse
Affiliation(s)
- Camilo Montero
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia; Translational Investigation Group, Sanitas University, Clinica Colsanitas, Bogota, Colombia.
| | - Nancy Yomayusa
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia; Translational Investigation Group, Sanitas University, Clinica Colsanitas, Bogota, Colombia; Infectious Diseases Department, Clinica Colsanitas, Bogota, Colombia
| | - Rodolfo Torres
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia; Translational Investigation Group, Sanitas University, Clinica Colsanitas, Bogota, Colombia
| | - Jorge Cortes
- Facultad de Medicina, Universidad Nacional de Colombia
| | - Carlos Alvarez
- Translational Investigation Group, Sanitas University, Clinica Colsanitas, Bogota, Colombia; Infectious Diseases Department, Clinica Colsanitas, Bogota, Colombia
| | - Juan Gallo
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia
| | - Guillermo Aldana
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia
| | - Andres Acevedo
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia; Translational Investigation Group, Sanitas University, Clinica Colsanitas, Bogota, Colombia
| | - Maria Rios
- Molecular Biology and Immunology Laboratory, Clinica Colsanitas, Bogota, Colombia
| | - Johana Echeverri
- Molecular Biology and Immunology Laboratory, Clinica Colsanitas, Bogota, Colombia
| | - Zuly Yepes
- Molecular Biology and Immunology Laboratory, Clinica Colsanitas, Bogota, Colombia
| | - Adriana Silva
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia
| | - Diana Gayon
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia
| | - Jorge Perez
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia; Translational Investigation Group, Sanitas University, Clinica Colsanitas, Bogota, Colombia; Infectious Diseases Department, Clinica Colsanitas, Bogota, Colombia; Molecular Biology and Immunology Laboratory, Clinica Colsanitas, Bogota, Colombia
| | - Milciades Ibanez
- Renal Transplantation Group, Clinica Reina Sofia, University Clinic, Bogota, Colombia; Translational Investigation Group, Sanitas University, Clinica Colsanitas, Bogota, Colombia
| |
Collapse
|
31
|
Geris JM, Spector LG, Pfeiffer RM, Limaye AP, Yu KJ, Engels EA. Cancer risk associated with cytomegalovirus infection among solid organ transplant recipients in the United States. Cancer 2022; 128:3985-3994. [PMID: 36126024 PMCID: PMC9633408 DOI: 10.1002/cncr.34462] [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: 04/04/2022] [Revised: 07/27/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) is among the most common viral infections after solid organ transplantation (SOT). Associations of CMV with cancer risk among SOT recipients have been incompletely evaluated. METHODS The authors used linked data from the US SOT registry and 32 cancer registries. Poisson regression was used to compare cancer incidence across CMV risk groups based on donor (D) and recipient (R) immunoglobulin G (IgG) serostatus: high risk (R-negative/D-positive), moderate risk (R-positive), and low risk (R-negative/D-negative). RESULTS In total, 247,318 SOT recipients were evaluated during 2000-2017 (R-negative/D-positive, 20.3%; R-positive, 62.9%; R-negative/D-negative, 16.8%). CMV-seropositive recipients were older, more racially/ethnically diverse, and had lower socioeconomic status than CMV-seronegative recipients. Compared with R-negative/D-negative recipients, recipients in the R-negative/D-positive and R-positive groups had a lower incidence of diffuse large B-cell lymphoma (DLBCL; R-negative/D-positive: adjusted incidence rate ratio [aIRR], 0.74; 95% confidence interval [CI], 0.59-0.91; R-positive: aIRR, 0.83; 95% CI, 0.69-1.00). CMV serostatus modified the association between Epstein-Barr virus (EBV) status and DLBCL (p = .0006): DLBCL incidence was increased for EBV R-negative/D-positive recipients (aIRR, 3.46; 95% CI, 1.50-7.95) among CMV R-negative/D-negative recipients but not among the other CMV risk groups. Compared with recipients who were CMV R-negative/D-negative, those who were R-negative/D-positive had a lower incidence of small intestine cancer (aIRR, 0.23; 95% CI, 0.09-0.63), and R-positive recipients had a higher incidence of lung cancer (aIRR, 1.24; 95% CI, 1.05-1.46). CMV status was not associated with risk for other cancers. CONCLUSIONS CMV status was not associated with risk for most cancers among SOT recipients. The inverse association with DLBCL may reflect the protective effects of CMV prophylaxis or treatment with off-target efficacy against EBV infection (the major cause of lymphoma in SOT recipients).
Collapse
Affiliation(s)
- Jennifer M. Geris
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis MN, USA
| | - Logan G. Spector
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Ruth M. Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda MD, USA
| | - Ajit P. Limaye
- Division of Allergy & Infectious Diseases, Department of Medicine, University of Washington, Seattle WA, USA
| | - Kelly J. Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda MD, USA
| | - Eric A. Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda MD, USA
| |
Collapse
|
32
|
Ouellette CP. Adoptive Immunotherapy for Prophylaxis and Treatment of Cytomegalovirus Infection. Viruses 2022; 14:v14112370. [PMID: 36366468 PMCID: PMC9694397 DOI: 10.3390/v14112370] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 01/31/2023] Open
Abstract
Cytomegalovirus (CMV), a member of the Herpesviridae family, is frequent among hematopoietic cell transplant (HCT) and solid organ transplant (SOT) recipients in absence of antiviral prophylaxis, and is a major cause of morbidity and mortality in these vulnerable populations. Antivirals such ganciclovir, valganciclovir, and foscarnet are the backbone therapies, however drug toxicity and antiviral resistance may render these agents suboptimal in treatment. Newer therapies such as letermovir and maribavir have offered additional approaches for antiviral prophylaxis as well as treatment of drug resistant CMV infection, though may be limited by cost, drug intolerance, or toxicity. Adoptive immunotherapy, the transfer of viral specific T-cells (VSTs), offers a new approach in treatment of drug-resistant or refractory viral infections, with early clinical trials showing promise with respect to efficacy and safety. In this review, we will discuss some of the encouraging results and challenges of widespread adoption of VSTs in care of immunocompromised patients, with an emphasis on the clinical outcomes for treatment and prophylaxis of CMV infection among high-risk patient populations.
Collapse
Affiliation(s)
- Christopher P Ouellette
- Division of Pediatric Infectious Diseases and Host Defense Program, Nationwide Children's Hospital, Columbus, OH 43205, USA
| |
Collapse
|
33
|
Zdziarski P, Gamian A. High Monocyte Count Associated with Human Cytomegalovirus Replication In Vivo and Glucocorticoid Therapy May Be a Hallmark of Disease. Int J Mol Sci 2022; 23:ijms23179595. [PMID: 36076989 PMCID: PMC9455616 DOI: 10.3390/ijms23179595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022] Open
Abstract
Cytomegalovirus (CMV) syndrome and infectious disease are defined as pathogen detection with appropriate clinical symptoms, but there are not pathognomonic signs of CMV disease. Although the prodrome of acute minor viral infections leukopenia (lymphopenia and neutropenia) is noted with onset of fever, followed by monocytosis, the role of monocytosis in CMV disease has not been described. Furthermore, under influence of corticosteroid therapy, CMV reactivation and monocytosis are described, but without a strict relationship with steroids dose. In the study, the monocyte level was investigated during the CMV infectious process. Regrettably, a non-selected group of 160 patients with high CMV viremia showed high dispersion of monocyte level and comparable with the median value for healthy subjects. Therefore, we investigated monocyte level in CMV-infected patients in relation to the logarithmic phase of the infectious process. Samples from patients with active CMV replication (exponential growth of CMV viremia) were tested. Significant monocytosis (above 1200/µL) during the logarithmic phase of CMV infection (with exponent between 3.23 and 5.77) was observed. Increased count and percentage of monocytes correlated with viral replication in several clinical situations except when there was a rapid recovery without relapse. Furthermore, glucocorticoids equivalent to 10 and 20 mg of dexamethasone during a 2–3-week period caused monocytosis—significant increase (to 1604 and 2214/µL, respectively). Conclusion: In light of the logarithmic increase of viral load, high monocytosis is a hallmark of CMV replication. In the COVID-19 era, presence of high virus level, especially part of virome (CMV) in the molecular technique, is not sufficient for the definition of either proven or probable CMV replication at any site. These preliminary observations merit additional studies to establish whether this clinical response is mediated by monocyte production or by decrease of differentiation to macrophages.
Collapse
Affiliation(s)
- Przemyslaw Zdziarski
- Lower Silesian Oncology, Pulmonology and Hematology Center, P.O. Box 1818, 50-385 Wroclaw, Poland
- Correspondence:
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| |
Collapse
|
34
|
Srivastava A, Bagchi S, Singh S, Balloni V, Agarwal SK. Assessment of Risk Factors and Outcome of Early Versus Late Cytomegalovirus Infection Infection in Living-related D+/R + Renal Allograft Recipients. Indian J Nephrol 2022; 32:47-53. [PMID: 35283573 PMCID: PMC8916147 DOI: 10.4103/ijn.ijn_463_20] [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: 09/28/2020] [Revised: 12/09/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Cytomegalovirus infection (CMV) in a kidney transplant recipient (KTR) is a serious complication resulting in increased morbidity, mortality and reduced graft survival. There is limited data on early (within 3 months posttransplant) CMV infection (ECMVI) vs. late CMV infection (LCMVI) in patients not receiving CMV prophylaxis. In India, majority of kidney transplants are D + R + combination. This study aimed to compare the risk factors and outcome of ECMVI vs. LCMVI in living related post-KTR. METHODS This was a single-center ambispective study of adult KTR from living donor between January 2001 and December 2015 who had CMV infection. This study had two cohorts: retrospective and prospective. Retrospective cohort included all KTR from January 2001 to September 2014. Prospective cohort included KTR who received transplants from October 2014 to December 2015. Of both cohorts, patients with early and late CMV infection were included. All patients received triple-drug immunosuppression. CMV infection was diagnosed when KTR had detectable CMV copies > 500/mL. In the prospective cohort, CMV PCR was done at 45 days, 3, 6, 9 and 12 months in all patients. Patients with CMV were treated on conventional lines. All patients were followed up till June 2016. RESULTS Of 2175 retrospective cohort, 97 and of the 155 prospective cohorts 75 had CMV infection, total being 172 CMV infections. Of these, 90 patients had ECNVI and 82 LCMVI. Induction was used in 48.8% in ECMVI group vs. 35.3% in LCMVI group (p = 0.02). CNI toxicity was present prior to CMV infection in 15 (17.4%) in ECMVI as compared to 14 (17.9%) in LCMVI (P = 0.93). In the ECMVI, 6 (6.6%) had acute rejection as compared to 13 (15.8%) in the LCMVI (P = 0.05). While asymptomatic CMV infection was more common in early (63.3% vs 37.8%, P = 0.001), symptomatic CMV without tissue diagnosis was more common in late (54.8% vs. 31.1%, P = 0.002). Total duration of post-transplant follow-up was 22.8 ± 22.1 months in ECMVI as compared to 49.7 + 40.9 months in the LCMVI (P < 0.001). The serum creatinine at last follow-up was 1.9 ± 1.6 mg/dL in ECMVI group and 2.4 ± 2.0 mg/dL in LCMVI (P = 0.02). CONCLUSION In D+/R + living renal transplant recipients, without routine CMV prophylaxis, late CMV infection had more tissue invasive disease and is associated with inferior graft function on long-term follow-up.
Collapse
Affiliation(s)
- Atul Srivastava
- Department of Nephrology, Base Hospital, Delhi Cantt, New Delhi, India
| | | | | | - Veena Balloni
- Technologist, Department of Laboratory Medicine, AIIMS, New Delhi, India
| | - Sanjay Kumar Agarwal
- Department of Nephrology, AIIMS, New Delhi, India
- Address for correspondence: Dr. Sanjay Kumar Agarwal, Professor and Head, Department of Nephrology, AIIMS, New Delhi - 110 049, India. E-mail:
| |
Collapse
|
35
|
Makharia G, Mohta S, Sridharan S, Gopalakrishnan R, Prasad N, Bansal S. Diarrhea in solid organ transplant recipients in the South Asian Region - Expert group opinion for diagnosis and management. INDIAN JOURNAL OF TRANSPLANTATION 2022; 16:23. [DOI: 10.4103/ijot.ijot_79_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
36
|
Low dose thymoglobulin versus basiliximab in cytomegalovirus positive kidney transplant recipients: Effectiveness of preemptive cytomegalovirus modified strategy. Nefrologia 2021. [DOI: 10.1016/j.nefro.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
37
|
Vutien P, Perkins J, Biggins SW, Reyes J, Imlay H, Limaye AP. Association of Donor and Recipient Cytomegalovirus Serostatus on Graft and Patient Survival in Liver Transplant Recipients. Liver Transpl 2021; 27:1302-1311. [PMID: 33687777 PMCID: PMC9121742 DOI: 10.1002/lt.26045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/12/2021] [Accepted: 02/24/2021] [Indexed: 01/03/2023]
Abstract
Among solid organ transplant recipients, donor cytomegalovirus (CMV) seropositive (D+) and recipient seronegative (R-) status are associated with an increased risk of graft loss and mortality after kidney or lung transplantation. Whether a similar relationship exists among liver transplant recipients (LTR) is unknown. We assessed graft loss and mortality among adult LTRs from January 1, 2010, to March 14, 2020, in the Organ Procurement and Transplantation Network database. We used multivariable mixed Cox proportional hazards regression to analyze the association of donor and recipient CMV serostatus group with graft loss and mortality, with donor seronegative (D-) and recipient seronegative (R-) as the reference group. Among 54,078 LTRs, the proportion of D-R-, D- and recipient seropositive (R+), D+R-, and D+R+ was 13.4%, 22.5%, 22%, and 42%, respectively. By unadjusted Kaplan-Meier survival curve estimates, survival by the end of follow-up was 73.3%, 73.5%, 70.1%, and 69.7%, among the D-R-, D-R+, D+R-, and D+R+ groups, respectively. By multivariable Cox regression, the CMV D+R- serogroup, but not other serogroups, was independently associated with increased risks of graft loss (adjusted hazard ratio [aHR], 1.13; 95% confidence interval [CI], 1.05-1.22) and mortality (aHR, 1.13; 95% CI, 1.05-1.22). The magnitude of the association of the CMV D+R- serostatus group with mortality was similar when the Cox regression analysis was restricted to the first year after transplant and beyond the first year after transplant: aHR, 1.13 (95% CI, 1.01-1.27) and aHR, 1.13 (95% CI, 1.02-1.25), respectively. Even in an era of CMV preventive strategies, CMV D+R- serogroup status remains independently associated with increased graft loss and mortality in adult LTRs. Factors in addition to direct CMV-associated short-term mortality are likely, and studies to define the underlying mechanism(s) are warranted.
Collapse
Affiliation(s)
- Philip Vutien
- Division of Gastroenterology and Hepatology, Center for Liver Investigation Fostering discovEry (C-LIFE), University of Washington, Seattle, WA
- Division of Transplant Surgery, Clinical and Bio-Analytics Transplant Laboratory University of Washington, Seattle, WA
| | - James Perkins
- Division of Transplant Surgery, Clinical and Bio-Analytics Transplant Laboratory University of Washington, Seattle, WA
- Division of Transplant Surgery, University of Washington, Seattle, WA
| | - Scott W. Biggins
- Division of Gastroenterology and Hepatology, Center for Liver Investigation Fostering discovEry (C-LIFE), University of Washington, Seattle, WA
- Division of Transplant Surgery, Clinical and Bio-Analytics Transplant Laboratory University of Washington, Seattle, WA
| | - Jorge Reyes
- Division of Transplant Surgery, Clinical and Bio-Analytics Transplant Laboratory University of Washington, Seattle, WA
- Division of Transplant Surgery, University of Washington, Seattle, WA
| | - Hannah Imlay
- Division of Infectious Disease, University of Utah, Salt Lake City, UT
| | - Ajit P. Limaye
- Division of Transplant Surgery, Clinical and Bio-Analytics Transplant Laboratory University of Washington, Seattle, WA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA
| |
Collapse
|
38
|
Affiliation(s)
- Fahad Aziz
- Division of Nephrology, Department of Medicine, University of Wisconsin–Madison School of Medicine and Public Health, University of Wisconsin Hospital and Clinics, Madison, Wisconsin
| | - Arjang Djamali
- Division of Nephrology, Department of Medicine, University of Wisconsin–Madison School of Medicine and Public Health, University of Wisconsin Hospital and Clinics, Madison, Wisconsin,Division of Transplantation, Department of Surgery, University of Wisconsin–Madison School of Medicine and Public Health, University of Wisconsin Hospital and Clinics, Madison, Wisconsin
| |
Collapse
|
39
|
Awad SM, Ali SM, Mansour YE, Fatahala SS. Synthesis and Evaluation of Some Uracil Nucleosides as Promising Anti-Herpes Simplex Virus 1 Agents. Molecules 2021; 26:2988. [PMID: 34069874 PMCID: PMC8157375 DOI: 10.3390/molecules26102988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Since herpes simplex virus type 1 (HSV-1) infection is so widespread, several antiviral drugs have been developed to treat it, among which are uracil nucleosides. However, there are major problems with the current medications such as severe side-effects and drug resistance. Here we present some newly synthesized cyclic and acyclic uracil nucleosides that showed very promising activity against HSV-1 compared to acyclovir.
Collapse
Affiliation(s)
- Samir Mohamed Awad
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo 11795, Egypt; (S.M.A.); (Y.E.M.)
| | - Shima Mahmoud Ali
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo 11795, Egypt;
| | - Yara Essam Mansour
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo 11795, Egypt; (S.M.A.); (Y.E.M.)
| | - Samar Said Fatahala
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Ain-Helwan, Cairo 11795, Egypt; (S.M.A.); (Y.E.M.)
| |
Collapse
|
40
|
Testa G, Johannesson L. The Value of a Nulliparous Uterus. Transplantation 2021; 105:958-959. [PMID: 32541560 DOI: 10.1097/tp.0000000000003347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Giuliano Testa
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX
| | | |
Collapse
|
41
|
Odler B, Deak AT, Pregartner G, Riedl R, Bozic J, Trummer C, Prenner A, Söllinger L, Krall M, Höflechner L, Hebesberger C, Boxler MS, Berghold A, Schemmer P, Pilz S, Rosenkranz AR. Hypomagnesemia Is a Risk Factor for Infections after Kidney Transplantation: A Retrospective Cohort Analysis. Nutrients 2021; 13:1296. [PMID: 33919913 PMCID: PMC8070921 DOI: 10.3390/nu13041296] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Magnesium (Mg2+) deficiency is a common finding in the early phase after kidney transplantation (KT) and has been linked to immune dysfunction and infections. Data on the association of hypomagnesemia and the rate of infections in kidney transplant recipients (KTRs) are sparse. METHODS We conducted a single-center retrospective cohort study of KTRs transplanted between 2005 and 2015. Laboratory data, including serum Mg2+ (median time of the Mg2+ measurement from KT: 29 days), rate of infections including mainly urinary tract infections (UTI), and common transplant-related viral infections (CMV, polyoma, EBV) in the early phase after KT were recorded. The primary outcome was the incidence of infections within one year after KT, while secondary outcomes were hospitalization due to infection, incidence rates of long-term (up to two years) infections, and all-cause mortality. RESULTS We enrolled 376 KTRs of whom 229 patients (60.9%) suffered from Mg2+ deficiency defined as a serum Mg2+ < 0.7 mmol/L. A significantly higher incidence rate of UTIs and viral infections was observed in patients with versus without Mg2+ deficiency during the first year after KT (58.5% vs. 47.6%, p = 0.039 and 69.9% vs. 51.7%, p < 0.001). After adjustment for potential confounders, serum Mg2+ deficiency remained an independent predictor of both UTIs and viral infections (odds ratio (OR): 1.73, 95% CI: 1.04-2.86, p = 0.035 and OR: 2.05, 95% CI: 1.23-3.41, p = 0.006). No group differences according to Mg2+ status in hospitalizations due to infections and infection incidence rates in the 12-24 months post-transplant were observed. In the Cox regression analysis, Mg2+ deficiency was not significantly associated with all-cause mortality (HR: 1.15, 95% CI: 0.70-1.89, p = 0.577). CONCLUSIONS KTRs suffering from Mg2+ deficiency are at increased risk of UTIs and viral infections in the first year after KT. Interventional studies investigating the effect of Mg2+ supplementation on Mg2+ deficiency and viral infections in KTRs are needed.
Collapse
Affiliation(s)
- Balazs Odler
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
- Transplant Center Graz, Medical University of Graz, A-8036 Graz, Austria;
| | - Andras T. Deak
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
- Transplant Center Graz, Medical University of Graz, A-8036 Graz, Austria;
| | - Gudrun Pregartner
- Institute of Medical Informatics, Statistics and Documentation, Medical University of Graz, A-8036 Graz, Austria; (G.P.); (R.R.); (A.B.)
| | - Regina Riedl
- Institute of Medical Informatics, Statistics and Documentation, Medical University of Graz, A-8036 Graz, Austria; (G.P.); (R.R.); (A.B.)
| | - Jasmin Bozic
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
| | - Christian Trummer
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (C.T.); (S.P.)
| | - Anna Prenner
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
- Transplant Center Graz, Medical University of Graz, A-8036 Graz, Austria;
| | - Lukas Söllinger
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
| | - Marcell Krall
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
| | - Lukas Höflechner
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
| | - Carina Hebesberger
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
- Transplant Center Graz, Medical University of Graz, A-8036 Graz, Austria;
| | - Matias S. Boxler
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
| | - Andrea Berghold
- Institute of Medical Informatics, Statistics and Documentation, Medical University of Graz, A-8036 Graz, Austria; (G.P.); (R.R.); (A.B.)
| | - Peter Schemmer
- Transplant Center Graz, Medical University of Graz, A-8036 Graz, Austria;
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, A-8036 Graz, Austria
| | - Stefan Pilz
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (C.T.); (S.P.)
| | - Alexander R. Rosenkranz
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria; (B.O.); (A.T.D.); (J.B.); (A.P.); (L.S.); (M.K.); (L.H.); (C.H.); (M.S.B.)
- Transplant Center Graz, Medical University of Graz, A-8036 Graz, Austria;
| |
Collapse
|
42
|
Veit T, Pan M, Munker D, Arnold P, Dick A, Kunze S, Meiser B, Schneider C, Michel S, Zoller M, Böhm S, Walter J, Behr J, Kneidinger N, Kauke T. Association of CMV-specific T-cell immunity and risk of CMV infection in lung transplant recipients. Clin Transplant 2021; 35:e14294. [PMID: 33749938 DOI: 10.1111/ctr.14294] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Protecting against CMV infection and maintaining CMV in latent state are largely provided by CMV-specific T-cells in lung transplant recipients. The aim of the study was to assess whether a specific T-cell response is associated with the risk for CMV infection in seronegative patients who are at high risk for delayed CMV infection. METHODS All CMV-seronegative recipients (R-) from CMV-seropositive donors (D+) between January 2018 and April 2019 were included and retrospectively screened for CMV infection before and after assessment of CMV-specific cell-mediated immunity. RESULTS Thirty-one of the 50 patients (62%) developed early-onset CMV infection. Lower absolute neutrophil counts were significantly associated with early-onset CMV infection. Antiviral prophylaxis was ceased after 137.2 ± 42.8 days. CMV-CMI were measured at a median of 5.5 months after LTx. 19 patients experienced early and late-onset CMV infection after prophylaxis withdrawal within 15 months post transplantation. Positive CMV-CMI was significantly associated with lower risk of late-onset CMV infection after transplantation in logistic and cox-regression analysis (OR=0.05, p = .01; OR=2,369, p = .026). CONCLUSION D+/R- lung transplant recipients are at high risk of developing early and late-onset CMV infection. Measurement of CMV-CMI soon after transplantation might further define the CMV infection prediction risk in LTx recipients being at high risk for CMV viremia.
Collapse
Affiliation(s)
- Tobias Veit
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany
| | - Ming Pan
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Laboratory for Immunogenetics, University of Munich, LMU, Munich, Germany
| | - Dieter Munker
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany
| | - Paola Arnold
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany
| | - Andrea Dick
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Laboratory for Immunogenetics, University of Munich, LMU, Munich, Germany
| | - Susanne Kunze
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Laboratory for Immunogenetics, University of Munich, LMU, Munich, Germany
| | - Bruno Meiser
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Transplant Center, University of Munich, LMU, Munich, Germany
| | - Christian Schneider
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Department of Thoracic Surgery, University of Munich, LMU, Munich, Germany
| | - Sebastian Michel
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Clinic of Cardiac Surgery, University of Munich, LMU, Munich, Germany
| | - Michael Zoller
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Department of Anaesthesiology, University of Munich, LMU, Munich, Germany
| | - Stephan Böhm
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Faculty of Medicine, Virology, Max von Pettenkofer Institute, University of Munich, LMU, Munich, Germany
| | - Julia Walter
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Department of Thoracic Surgery, University of Munich, LMU, Munich, Germany
| | - Jürgen Behr
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany
| | - Nikolaus Kneidinger
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany
| | - Teresa Kauke
- Department of Medicine V, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), University Hospital, LMU Munich, Munich, Germany.,Laboratory for Immunogenetics, University of Munich, LMU, Munich, Germany.,Department of Thoracic Surgery, University of Munich, LMU, Munich, Germany
| |
Collapse
|
43
|
Predictive factors of human cytomegalovirus reactivation in newly diagnosed glioblastoma patients treated with chemoradiotherapy. J Neurovirol 2021; 27:94-100. [PMID: 33405205 DOI: 10.1007/s13365-020-00922-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 09/16/2020] [Accepted: 10/08/2020] [Indexed: 12/31/2022]
Abstract
The human cytomegalovirus (HCMV) is a ubiquitous herpes virus which infects 40 to 99% of the population. HCMV reactivation may occur in the context of immunosuppression and can induce significant morbidities. Several cases of HCMV infections or HCMV reactivation have thus been reported in glioblastoma (GBM) patients treated with radio(chemo)therapy. With the aim to identify the main risk factors associated with HCMV reactivation, we reviewed all patients treated for a newly diagnosed GBM in our institution from October 2013 to December 2015. Age, sex, Karnofsky performance status (KPS), absolute lymphocyte count (ALC), serological HCMV status, and steroid doses were recorded at the start and 1 month after the end of radiotherapy (RT). Within the 103 patients analyzed, 34 patients (33%) had an initial negative serology for HCMV, and none of them developed a seroconversion after treatment. Among patients with positive HCMV IgG (n = 69), 16 patients (23%) developed a viremia at one point during treatment. Age (> 60 years), steroid intake, and ALC (< 1500/mm3) before RT were correlated with HCMV reactivation. HCMV viremia was associated with neurological decline 1 month after chemoradiotherapy but progression-free survival was not impacted. A shorter overall survival was seen in these patients when compared with the others, but this could be biased by the older age in this subgroup. HCMV reactivation needs to be sought in case of a neurological decline during RT especially in older patients treated with steroids and low lymphocytes counts.
Collapse
|
44
|
Cytomegalovirus disease in de novo kidney-transplant recipients: comparison of everolimus-based immunosuppression without prophylaxis with mycophenolic acid-based immunosuppression with prophylaxis. Int Urol Nephrol 2020; 53:591-600. [PMID: 33058036 DOI: 10.1007/s11255-020-02676-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/03/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To compare everolimus (EVR) plus low-dose tacrolimus (TAC) with mycophenolic acid (MPA) plus standard-dose TAC with regards to rates of cytomegalovirus (CMV) disease in de novo kidney-transplant recipients (KTRs). METHODS This single-center retrospective study included 187 de novo KTRs; 59 patients (31.6%) received EVR/low-dose TAC (group 1); 128 patients (68.4%) received MPA with standard-dose TAC (group 2). All received anti-thymocyte globulins as the induction therapy, and steroid-sparing strategy. Valganciclovir prophylaxis was mandatory for CMV D+/R- KTRs (seronegative recipients of a seropositive donor) in both groups and for R+ seropositive recipients (only in group 2). RESULTS The 2-year incidence of CMV disease was low and comparable between groups: 6.8% and 7.0% in groups 1 and 2, respectively (p = 0.94). There was no statistical difference in CMV serostatus (p = 1). However, CMV disease tended to be less frequent, though not statistically different, in R+ KTRs receiving EVR without prophylaxis (3.7% vs. 8.5% in groups 1 and 2, respectively) and in patients without EVR discontinuation (2.6% vs. 6.9% in patients who did not discontinue MPA (p = 0.29). Two-year graft function was good and comparable between groups (median eGFR of 54.2 and 53.0 mL/min in groups 1 and 2, respectively; p = 0.47); incidence of immunological events was low. Significantly more patients in group 1 discontinued EVR because of adverse events than patients that discontinued MPA in group 2 (35.6% in group 1 vs. 10.2% in group 2; p < 0.001). CONCLUSIONS Everolimus plus low-dose TAC given to de novo KTRs was associated with low rates of CMV disease, especially in R+ patients with no CMV prophylaxis.
Collapse
|
45
|
Lizaola-Mayo BC, Rodriguez EA. Cytomegalovirus infection after liver transplantation. World J Transplant 2020; 10:183-190. [PMID: 32844094 PMCID: PMC7416364 DOI: 10.5500/wjt.v10.i7.183] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/28/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (CMV) represents the most common opportunistic infection in liver transplant recipients. CMV infections in post liver transplant patients cause significant morbidity and mortality, directly affecting post-transplant outcomes. This review will provide the framework for the surveillance, diagnosis, prophylaxis and treatment of CMV in the liver transplant population.
Collapse
Affiliation(s)
- Blanca C Lizaola-Mayo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Phoenix, AZ 85259, United States
| | - Eduardo A Rodriguez
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah, Salt Lake City, UT 84132, United States
| |
Collapse
|
46
|
Efficacy and Safety of a Weight-based Dosing Regimen of Valganciclovir for Cytomegalovirus Prophylaxis in Pediatric Solid-organ Transplant Recipients. Transplantation 2020; 103:1730-1735. [PMID: 31343571 DOI: 10.1097/tp.0000000000002632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Valganciclovir has been widely used for cytomegalovirus (CMV) prophylaxis in solid-organ transplant recipients. However, the optimal dosing protocol and target exposure in children are still unclear. Specific data as to the efficacy and safety of low-dose/low-exposure regimens are lacking and urgently needed. METHODS During 2010 to 2015, the clinical efficacy and safety of a weight-based regimen of valganciclovir of 17 mg/kg/day, with a stratified dose reduction for impaired creatinine clearance, given as a CMV prophylaxis for 3 to 6 months, was retrospectively evaluated among pediatric kidney and liver transplant recipients, 12 months posttransplantation. Incidence of CMV infection was assessed by periodic measurements of viral load; adverse events were evaluated. RESULTS Eighty-three children who had undergone 86 transplantations and were treated with 17 mg/kg of valganciclovir were included. Median age was 9.77 years (range, 0.6 to 18.9). Twelve (14%) developed CMV infection: 1 during prophylaxis and 11 during follow-up. These events comprised 6 cases of asymptomatic viremia and 6 cases of a clinically significant disease without occurrences of tissue-invasive disease. Treatment-related adverse effects occurred in 7 patients (8%), mostly hematological, resulting in premature drug cessation. CONCLUSIONS Our results support the use of 17 mg/kg of valganciclovir for CMV prophylaxis in liver and kidney transplanted children as it showed satisfactory long-term efficacy and a good safety profile.
Collapse
|
47
|
Lim EY, Jackson SE, Wills MR. The CD4+ T Cell Response to Human Cytomegalovirus in Healthy and Immunocompromised People. Front Cell Infect Microbiol 2020; 10:202. [PMID: 32509591 PMCID: PMC7248300 DOI: 10.3389/fcimb.2020.00202] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/16/2020] [Indexed: 12/16/2022] Open
Abstract
While CD8+ T cells specific for human cytomegalovirus (HCMV) have been extensively studied in both healthy HCMV seropositive carriers and patients undergoing immunosuppression, studies on the CD4+ T cell response to HCMV had lagged behind. However, over the last few years there has been a significant advance in our understanding of the importance and contribution that CMV-specific CD4+ T cells make, not only to anti-viral immunity but also in the potential maintenance of latently infected cells. During primary infection with HCMV in adults, CD4+ T cells are important for the resolution of symptomatic disease, while persistent shedding of HCMV into urine and saliva is associated with a lack of HCMV specific CD4+ T cell response in young children. In immunosuppressed solid organ transplant recipients, a delayed appearance of HCMV-specific CD4+ T cells is associated with prolonged viremia and more severe clinical disease, while in haematopoietic stem cell transplant recipients, it has been suggested that HCMV-specific CD4+ T cells are required for HCMV-specific CD8+ T cells to exert their anti-viral effects. In addition, adoptive T-cell immunotherapy in transplant patients has shown that the presence of HCMV-specific CD4+ T cells is required for the maintenance of HCMV-specific CD8+ T cells. HCMV is a paradigm for immune evasion. The presence of viral genes that down-regulate MHC class II molecules and the expression of viral IL-10 both limit antigen presentation to CD4+ T cells, underlining the important role that this T cell subset has in antiviral immunity. This review will discuss the antigen specificity, effector function, phenotype and direct anti-viral properties of HCMV specific CD4+ T cells, as well as reviewing our understanding of the importance of this T cell subset in primary infection and long-term carriage in healthy individuals. In addition, their role and importance in congenital HCMV infection and during immunosuppression in both solid organ and haemopoietic stem cell transplantation is considered.
Collapse
Affiliation(s)
| | | | - Mark R. Wills
- Division of Infectious Diseases, Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
48
|
Where do we Stand after Decades of Studying Human Cytomegalovirus? Microorganisms 2020; 8:microorganisms8050685. [PMID: 32397070 PMCID: PMC7284540 DOI: 10.3390/microorganisms8050685] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 12/26/2022] Open
Abstract
Human cytomegalovirus (HCMV), a linear double-stranded DNA betaherpesvirus belonging to the family of Herpesviridae, is characterized by widespread seroprevalence, ranging between 56% and 94%, strictly dependent on the socioeconomic background of the country being considered. Typically, HCMV causes asymptomatic infection in the immunocompetent population, while in immunocompromised individuals or when transmitted vertically from the mother to the fetus it leads to systemic disease with severe complications and high mortality rate. Following primary infection, HCMV establishes a state of latency primarily in myeloid cells, from which it can be reactivated by various inflammatory stimuli. Several studies have shown that HCMV, despite being a DNA virus, is highly prone to genetic variability that strongly influences its replication and dissemination rates as well as cellular tropism. In this scenario, the few currently available drugs for the treatment of HCMV infections are characterized by high toxicity, poor oral bioavailability, and emerging resistance. Here, we review past and current literature that has greatly advanced our understanding of the biology and genetics of HCMV, stressing the urgent need for innovative and safe anti-HCMV therapies and effective vaccines to treat and prevent HCMV infections, particularly in vulnerable populations.
Collapse
|
49
|
Uchida H, Sakamoto S, Shimizu S, Takeda M, Yanagi Y, Fukuda A, Uchiyama T, Irie R, Kasahara M. Efficacy of Antithymocyte Globulin Treatment for Severe Centrilobular Injury Following Pediatric Liver Transplant: Clinical Significance of Monitoring Lymphocyte Subset. EXP CLIN TRANSPLANT 2020; 18:325-333. [PMID: 32281527 DOI: 10.6002/ect.2019.0387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Central perivenulitis can occur in association with T-cell-mediated rejection and can sometimes require strong immunosuppressant therapy as refractory rejection. Furthermore, patients with central perivenulitis are more likely to have subsequent episodes of T-cell-mediated rejection and develop chronic rejection than those without central perivenulitis. We retrospectively analyzed clinical data of pediatric patients with episodes of T-cell-mediated rejection according to severity of central perivenulitis and monitored HLA-DR-positive CD8-positive T cells and recent thymic emigrants during treatment for T-cell-mediated rejection. MATERIALS AND METHODS We identified biopsy-proven T-cell-mediated rejection in 50 liver transplant recipients (45 with living-related donors, 5 with deceased donors) between September 2014 and August 2018. Lymphocyte subsets in peripheral blood samples were analyzed. RESULTS Of 50 pediatric patients, 30 were boys and 20 were girls (median age at transplant of 1.1 y; interquartile range, 0.6-6.2 y). Central perivenulitis was found in 46 patients (92%), which was mild in 13, moderate in 16, and severe in 17. Antithymocyte globulin was more frequently administered to patients with severe central perivenulitis than others (P < .05). Patients with antithymocyte globulin treatment were less likely to have subsequent episodes of T-cell-mediated rejection than those without this treatment (P < .05). The absolute number of CD8-positive HLA-DR-positive T cells in patients during treatment was significantly higher than in control patients (P < .05). The absolute number of recent thymic emigrants in patients with active infection was significantly lower than in patients without infection (P < .05). CONCLUSIONS Our results suggest the efficacy and safety of antithymocyte globulin for treating T-cell-mediated rejection with severe central perivenulitis in pediatric liver transplant recipients and suggest that antithymocyte globulin can prevent subsequent episodes of T-cell-mediated rejection. Analyzing lymphocyte subsets during treatment for rejection may help highlight viable therapeutic strategies for achieving a good outcome.
Collapse
Affiliation(s)
- Hajime Uchida
- From the Organ Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Herrera S, Khan B, Singer LG, Binnie M, Chaparro C, Chow CW, Martinu T, Tomlinson G, Keshavjee S, Husain S, Tikkanen JM. Extending cytomegalovirus prophylaxis in high-risk (D+/R-) lung transplant recipients from 6 to 9 months reduces cytomegalovirus disease: A retrospective study. Transpl Infect Dis 2020; 22:e13277. [PMID: 32170813 DOI: 10.1111/tid.13277] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 02/25/2020] [Accepted: 03/08/2020] [Indexed: 12/31/2022]
Abstract
RATIONALE Cytomegalovirus (CMV)-seronegative recipients receiving a seropositive allograft (D+/R-) are at a high risk of developing CMV disease. Our program increased the duration of CMV prophylaxis from 6 to 9 months in May 2013. Here, we present the impact on the incidence of CMV infection, disease, side effects, rejection, and other factors. METHODS Retrospective cohort of 241 CMV (D+/R-) patients transplanted between January 1, 2008, and December 31, 2017. Blood CMV testing was done according to protocol. All patients received ganciclovir/valganciclovir as prophylaxis. We compared the incidence and timing of CMV infection and disease up to 6 months after cessation of prophylaxis between patients who received 9 months (May 2013 onwards) and a historical control group who received 6 months of prophylaxis (prior to May 2013). CMV infection was defined as detectable CMV viremia in the absence of symptoms. CMV disease was defined as CMV syndrome or tissue-invasive disease. Side effects of prophylaxis and CMV resistance were recorded. RESULTS A total of 116 patients were included in the 6-month group and 125 in the 9-month group. The extended 9-month CMV prophylaxis delayed the onset of CMV infection (median time to CMV infection after lung transplantation 295 vs 353 days, P < .01) but did not significantly reduce the incidence of CMV infection (65% vs 64%, P = .06, log-rank). The 9-month prophylaxis delayed the onset and decreased the incidence of CMV disease from 50% in the 6-month group to 42% (P = .02 log-rank). There was no difference in the rate of adverse effects (leukopenia in 32% in both groups, P = .53) or development of CMV resistance between the two groups (4 cases in both groups, P = .92). There were no significant differences in overall survival or the rate of chronic lung allograft dysfunction between the groups. CONCLUSIONS Extending duration of CMV prophylaxis from 6 to 9 months resulted in a delayed and decreased incidence of CMV disease in our lung transplant population. The absolute risk reduction achieved by extended CMV prophylaxis was 8%. The incidence of CMV infection, and ganciclovir resistance and side effects were similar between the two groups. Our results suggest that extending CMV prophylaxis in the highest risk CMV D+/R- group is effective in reducing CMV disease.
Collapse
Affiliation(s)
- Sabina Herrera
- Transplant Infectious Diseases, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Basha Khan
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Lianne G Singer
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Matthew Binnie
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Cecilia Chaparro
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Chung-Wai Chow
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Tereza Martinu
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| | - George Tomlinson
- Department of Medicine, University Health Network / University of Toronto, Toronto, ON, Canada
| | - Shaf Keshavjee
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Shahid Husain
- Transplant Infectious Diseases, University Health Network/University of Toronto, Toronto, ON, Canada
| | - Jussi M Tikkanen
- Toronto Lung Transplant Program, University Health Network/University of Toronto, Toronto, ON, Canada
| |
Collapse
|