1
|
Hirosawa M, Nakanishi T, Tanaka A, Akao K, Higashi T, Morimoto H, Tsukada J. Viral and Fungal Infections Early After HLA-Mismatched Hematopoietic Stem Cell Transplantation Using Low-Dose Antithymocyte Globulin in High-Risk Patients With Hematological Malignancies Not in Remission. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e348-e358. [PMID: 39865001 DOI: 10.1016/j.clml.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND In vivo T-cell depletion with antithymocyte globulin (ATG), especially at high-doses has been shown to be associated with increased incidence of infections after allogeneic hematopoietic stem cell transplantation (HSCT). However, it remains unclear whether ATG, even at low-doses increases the risk of posttransplant infections in the high-risk HSCT setting. PATIENTS AND METHODS We conducted a single-center retrospective study of viral and fungal infections early after transplantation, using the data from 82 patients with hematological malignancies. Among them, 42 underwent HLA-mismatched HSCT using low-dose (2.5 mg/kg n = 41, 2.0 mg/kg n = 1) thymoglobulin (ATG patients), and 40 control patients received HSCT without ATG (non-ATG patients) during the same period. Cord blood transplantation patients were excluded. All ATG patients had hematological malignancies not in remission at the time of transplantation, and were considered to be at high-risk for posttransplant infections. RESULTS There were no appreciable between-group differences in the incidence of clinically significant cytomegalovirus infection (csCMVi), late-onset CMV reactivation after discontinuation of letermovir, invasive fungal diseases or Epstein-Barr virus (EBV)-associated posttransplant lymphoproliferative disease. Peak values of CMV antigenemia were almost equal in ATG and non-ATG patients. The prevention of csCMVi with letermovir was constant in the 2 groups. However, ATG patients showed earlier reactivation of CMV and higher incidence of EBV viremia than non-ATG patients. Among their underlying diseases, mature T-cell neoplasm was a significant risk factor for CMV/EBV reactivation. CONCLUSION The use of low-dose thymoglobulin in HLA-mismatched HSCT for nonremission hematological malignancies is a reasonable strategy under careful monitoring for viral reactivation.
Collapse
Affiliation(s)
- Makoto Hirosawa
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tsukasa Nakanishi
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Aya Tanaka
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kenichi Akao
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takehiro Higashi
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hiroaki Morimoto
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Junichi Tsukada
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu, Japan.
| |
Collapse
|
2
|
Leclerc V, Châteauvert N. Risk of Cytomegalovirus Infection after 3 or 6 Months of Valganciclovir Prophylaxis among Donor-Seropositive, Recipient-Seronegative Heart Transplant Recipients. Can J Hosp Pharm 2025; 78:e3701. [PMID: 40371188 PMCID: PMC12057816 DOI: 10.4212/cjhp.3701] [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: 08/15/2024] [Accepted: 11/11/2024] [Indexed: 05/16/2025]
Abstract
Background Cytomegalovirus (CMV) disease is frequent following heart transplant, especially among patients with donor-seropositive, recipient-seronegative (D+/R-) CMV status. Valganciclovir prophylaxis for 3 to 6 months is recommended, but few data support the use of 6 rather than 3 months of prophylaxis for patients who have undergone heart transplant. Objective To compare the risk of CMV infection and a first occurrence of CMV disease after heart transplant among CMV D+/R- patients who received 3 or 6 months of prophylaxis in the first year after transplant. Methods This retrospective analysis included every D+/R- heart transplant recipient at the study centre between October 2015 and October 2022. Patients had to have a minimum of 1 year of follow-up for inclusion. Results Thirty-five patients met the inclusion criteria. The duration of valganciclovir prophylaxis (determined by the medical team) was 3 months for 22 patients and 6 months for 13 patients. CMV infection occurred in 68.2% (15/22) and 30.8% (4/13) of the 3-month and 6-month groups, respectively (risk difference 37.4%, 95% confidence interval [CI] 1.75 to 65.8, p = 0.04). CMV disease occurred in 54.5% (12/22) of patients who received 3 months of prophylaxis and 23.1% (3/13) of those who received 6 months of prophylaxis (risk difference 31.4%, 95% CI -3.7 to 59.4, p = 0.09). Most CMV infections and diseases occurred in the 3 months after the end of prophylaxis. Conclusions After heart transplant, CMV D+/R- patients who received 3 months of prophylaxis had a higher risk of CMV infection and a non-statistically significant higher risk of CMV disease relative to those who received 6 months of prophylaxis.
Collapse
Affiliation(s)
- Vincent Leclerc
- , BPharm, MSc, is with the Département de pharmacie, Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, and the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, Quebec
| | - Nathalie Châteauvert
- , BSc, BPharm, MSc, is with the Département de pharmacie, Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, and the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec, Quebec
| |
Collapse
|
3
|
Lynch K, Märtson AG. The Importance of Drug Exposure in the Development of Cytomegalovirus Resistance. Int J Antimicrob Agents 2025:107537. [PMID: 40374080 DOI: 10.1016/j.ijantimicag.2025.107537] [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: 01/10/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
Human cytomegalovirus (CMV) is a widespread pathogen which remains asymptomatic in healthy individuals. However, CMV disease can be life-threatening in immunocompromised individuals, particularly in transplant patients. This disease is routinely managed by antiviral agents including (val)ganciclovir, foscarnet, cidofovir, letermovir, and maribavir. Subtherapeutic antiviral drug exposure is a common occurrence and can lead to drug-resistant CMV development, a key contributor to disease progression. Breakthrough CMV often results in graft loss, end-organ failure, or death. By optimising intracellular exposure levels of antiviral therapies, it may be possible to improve patient outcomes. Therefore, this review aims to explore the relationship between antiviral exposure and the development of drug-resistant CMV. There are several challenges to achieving optimal concentrations of current and novel CMV therapies. Narrow therapeutic indices and toxicity profiles of current CMV therapeutics contribute to their subtherapeutic exposure and hence suboptimal clinical outcomes. Alternately, novel antivirals such as letermovir and maribavir offer improved pharmacokinetic profiles. However, these agents are associated with rapid resistance development. Overall, a distinct gap exists in understanding the relationship between antiviral exposure and resistance development. As a result, current clinical markers used to predict clinical efficacy lack reliability. In future, resistance development in relation to drug exposure should be included as a clinical trial endpoint to gain understanding of exposure-resistance relationships. With solid knowledge of these relationships, more predictive in vitro and in vivo markers of clinical efficacy can be identified. Additionally, pharmacokinetic-pharmacodynamic models and combination therapies should be further explored in to improve the management of CMV.
Collapse
Affiliation(s)
- Katie Lynch
- Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, The Netherlands
| | - Anne-Grete Märtson
- Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, The Netherlands.
| |
Collapse
|
4
|
Huang JS, Wang HK, Rong LP, Jiang XY, Liu LS, Huang LY, Zhang N, Yue ZH. Case Report: Maribavir for refractory cytomegalovirus viremia after renal transplantation in a child with Schimke's immune-osseous dysplasia. Front Immunol 2025; 16:1521763. [PMID: 40260257 PMCID: PMC12009931 DOI: 10.3389/fimmu.2025.1521763] [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: 11/02/2024] [Accepted: 03/17/2025] [Indexed: 04/23/2025] Open
Abstract
Cytomegalovirus (CMV) is a major opportunistic pathogen in recipients of solid organ transplantation. Maribavir, a pUL97 protein kinase inhibitor, was approved for the treatment of refractory post-transplant CMV infection in the US in 2021. However, it is rarely used in pediatric patients worldwide. Here, we report the case of a Chinese boy with Schimke's immune-osseous dysplasia (SIOD) who developed refractory CMV infection after a renal transplantation. An 11-year-old boy was hospitalized with recurrent abdominal and testicular pain 50 days after renal transplantation. Diagnoses included urinary tract infection, epididymitis, CMV viremia, stage 2 chronic kidney disease, and SIOD. After five days of treatment, his pain improved, but he developed persistent fever and shortness of breath. Blood CMV levels rose to 1.64 × 105 copies/ml after one month of ganciclovir treatment. Significant bone marrow suppression was observed after combined treatment with foscarnet. Anti-rejection treatment was discontinued due to compromised immune function. On day 40, maribavir was initiated with parental consent, resulting in undetectable CMV copies within four days. The patient's clinical status and bone marrow suppression had improved. Continuing maribavir for two weeks led to the disappearance of CMV viremia, no bone marrow suppression, and normal liver and kidney functions. This case demonstrates the successful short-term use of maribavir in the treatment of refractory CMV infection in an immune-deficient child after renal transplantation. Further studies are required to explore the efficacy and safety of maribavir in pediatric patients.
Collapse
Affiliation(s)
- Jia-Shuan Huang
- Pediatrics Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Medical College, Sun Yat-Sen University, Guangzhou, China
| | - Hong-Kai Wang
- Medical College, Sun Yat-Sen University, Guangzhou, China
| | - Li-Ping Rong
- Pediatrics Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Yun Jiang
- Pediatrics Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Long-Shan Liu
- Organ Transplantation Department, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liu-Yi Huang
- Pediatrics Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Na Zhang
- Pediatrics Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhi-Hui Yue
- Pediatrics Department, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
5
|
Hinze CA, Simon S, Gottlieb J. Respiratory infections in lung transplant recipients. Curr Opin Infect Dis 2025; 38:150-160. [PMID: 39927477 DOI: 10.1097/qco.0000000000001097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
PURPOSE OF REVIEW Morbidity and mortality rates after lung transplantation still remain higher than after other forms of solid organ transplantation, primarily due to a higher risk of infections and the development of chronic lung allograft dysfunction. Thus, a tiered approach highlighting the most significant respiratory pathogens including common opportunistic infections along with diagnostic, treatment and prevention strategies, including vaccination and prophylaxis is needed. RECENT FINDINGS The need for intense immunosuppressive therapy to prevent rejection, coupled with the transplanted lung's constant exposure to environment and impaired local defence mechanisms leads to frequent infections. Viral and bacterial infections are most frequent while fungal infections mainly involve the tracheobronchial tract but may be fatal in case of disseminated disease. Some infectious agents are known to trigger acute rejection or contribute to chronic allograft dysfunction. Invasive testing in the form of bronchoscopy with bronchoalveolar lavage is standard and increasing experience in point of care testing is gained to allow early preemptive therapy. SUMMARY Timely diagnosis, treatment, and ongoing monitoring are essential, but this can be difficult due to the wide variety of potential pathogens.
Collapse
Affiliation(s)
- Christopher Alexander Hinze
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Susanne Simon
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School
| | - Jens Gottlieb
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
6
|
Guo Z, Zhu J, Wang J, Wang L, Tang F, Huang H, Xia Z, Liu L, Wang D, Zhong N, Zhou H, Zhou Z, Dai W, Xu X, Zhou H, Deng L, Meng J, Sun Z, Shao L, Cao YJ, Liu Y, Qu R, Li G, Chen P, Zhang H, Liang J, Li Y, Liu J, Xu Z, Sung Inda S, Xiang X, Wu Q, Wang Q. Chinese expert consensus on the application of intravenous immunoglobulin in hematological diseases. Front Med (Lausanne) 2025; 12:1544025. [PMID: 40236459 PMCID: PMC11996829 DOI: 10.3389/fmed.2025.1544025] [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: 12/12/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Intravenous immunoglobulin (IVIG), first developed for the treatment of patients with antibody deficiencies, is now widely used in clinical practice, especially in hematological and immune system diseases, and its application in hematological oncology chemotherapy, cellular immunotherapy and hematopoietic stem cell transplantation (HSCT) is becoming more and more common. The Chinese Collaborative Group for Infection Immunology and Microecology Research Translation Collaborative Group organized relevant experts to discuss and propose the "Chinese expert consensus on the application of intravenous immunoglobulin in hematological diseases," which was formulated based on the progress of research on the application of IVIG in blood diseases, and provides a basis for the standardization of the use of IVIG in hematologic disorders.
Collapse
Affiliation(s)
- Zhi Guo
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jie Zhu
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jun Wang
- Department of Hematology, Hongkong University Shenzhen Hospital, Shenzhen, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Feifei Tang
- Department of Hematology, Peking University People’s Hospital, Beijing, China
| | - Huiqiang Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhongjun Xia
- State Key Laboratory of Oncology in South China, Department of Hematology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Liqiong Liu
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Danyu Wang
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Nan Zhong
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Huanhuan Zhou
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhaogui Zhou
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Wei Dai
- Department of Hematology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hao Zhou
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijuan Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Jingye Meng
- Department of Hematology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Zhiqiang Sun
- Department of Hematology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Liang Shao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu J. Cao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yansong Liu
- Department of Critical Care Medicine, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Rong Qu
- Department of Critical Care Medicine, Huizhou Central People Hospital, Huizhou, China
| | - Guowei Li
- Department of Hematology, Huizhou Central People Hospital, Huizhou, China
| | - Peng Chen
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongyan Zhang
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing Liang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yuhua Li
- Hematology Department, Southern Medical University, Zhujiang Hospital, Guangzhou, China
- Guangdong Engineering Research Center of Precision Immune Cell Therapy Technology, Guangzhou, China
| | - Jiajun Liu
- Department of Hematology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zishan Xu
- Department of Hematology, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong SAR, China
| | - Soong Sung Inda
- Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong SAR, China
| | - Xiaochen Xiang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Qingming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Conesa L, Gonzalez-Silva G, Peris-Serra L, Garriga-Edo S, Castellote L, Ferrer R, Villena Y. Simultaneous Monitoring of 3 Antiviral Drugs in Serum Using Liquid Chromatography-Tandem Mass Spectrometry: Full Validation and Clinical Application. Ther Drug Monit 2025:00007691-990000000-00331. [PMID: 40100067 DOI: 10.1097/ftd.0000000000001321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Patients undergoing solid organ and hematopoietic stem cell transplantation are at risk of opportunistic pathogenic infections that increase morbidity and mortality. Universal antiviral prophylaxis improves the outcomes in this context. Therapeutic drug monitoring of antiviral drugs is not universally recommended but may be necessary in certain complex or polymorbid patients. The authors aimed to develop and validate a high-performance liquid chromatography-tandem mass spectrometry method to simultaneously quantify ganciclovir, acyclovir, and letermovir in human serum. METHODS A stable isotopically labeled internal standard was used for each antiviral drug. Compounds were extracted by protein precipitation, evaporation, and reconstitution in an aqueous mobile phase. Samples were analyzed using reverse-phase chromatography with subsequent detection by electrospray ionization in the positive ion mode on a triple quadrupole mass spectrometer (run time: 6.5 minutes). RESULTS Analytical curves for ganciclovir and acyclovir exhibited linearity within 0.1-25 mg/L (R2 > 0.993), whereas for letermovir, the linear range was 0.01-2 mg/L (R2 = 0.999). Matrix effects were not observed. Intraday and interday precision and accuracy were within ±15%. A therapeutic drug monitoring-guided strategy was explored to optimize preemptive antiviral drug therapy in 3 cohorts of transplant recipients. Seventy-nine samples from 35 patients were quantified, revealing median trough concentrations of 0.2 mg/L for ganciclovir (n = 21), 0.28 mg/L for acyclovir (n = 26), and 0.29 mg/L for letermovir (n = 32). CONCLUSIONS This method has been successfully applied in clinical settings and allows reliable and accurate drug-level measurements.
Collapse
Affiliation(s)
- Laura Conesa
- Clinical Biochemistry Department Vall D'Hebron University Hospital. Clinical Biochemistry, Drug Delivery and Therapy Research Group. Vall D'Hebrón Research Institute (VHIR), Vall D'Hebron Barcelona Hospital Campus, Barcelona, Spain; and
| | - Gonzalo Gonzalez-Silva
- Clinical Biochemistry Department Vall D'Hebron University Hospital. Clinical Biochemistry, Drug Delivery and Therapy Research Group. Vall D'Hebrón Research Institute (VHIR), Vall D'Hebron Barcelona Hospital Campus, Barcelona, Spain; and
| | - Lydia Peris-Serra
- Clinical Biochemistry Department Vall D'Hebron University Hospital. Clinical Biochemistry, Drug Delivery and Therapy Research Group. Vall D'Hebrón Research Institute (VHIR), Vall D'Hebron Barcelona Hospital Campus, Barcelona, Spain; and
| | - Sarai Garriga-Edo
- Clinical Biochemistry Department Vall D'Hebron University Hospital. Clinical Biochemistry, Drug Delivery and Therapy Research Group. Vall D'Hebrón Research Institute (VHIR), Vall D'Hebron Barcelona Hospital Campus, Barcelona, Spain; and
| | - Laura Castellote
- Clinical Biochemistry Department Vall D'Hebron University Hospital. Clinical Biochemistry, Drug Delivery and Therapy Research Group. Vall D'Hebrón Research Institute (VHIR), Vall D'Hebron Barcelona Hospital Campus, Barcelona, Spain; and
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Roser Ferrer
- Clinical Biochemistry Department Vall D'Hebron University Hospital. Clinical Biochemistry, Drug Delivery and Therapy Research Group. Vall D'Hebrón Research Institute (VHIR), Vall D'Hebron Barcelona Hospital Campus, Barcelona, Spain; and
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Yolanda Villena
- Clinical Biochemistry Department Vall D'Hebron University Hospital. Clinical Biochemistry, Drug Delivery and Therapy Research Group. Vall D'Hebrón Research Institute (VHIR), Vall D'Hebron Barcelona Hospital Campus, Barcelona, Spain; and
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
8
|
Potluri VS, Goldberg D, Zhang S, Schaubel DE, Molnar MZ, Forbes R, Sise ME, Rogers JL, Balaraman V, Bhalla A, Shaffer D, Concepcion BP, Chung RT, Strohbehn IA, Mapchan S, Vujjini V, Sangadi A, Martin E, Bloom RD, Ammazzalorso A, Blumberg EA, Reese PP. Risk of Cytomegalovirus Viremia Following Transplantation of Hepatitis C-Viremic Donor Kidneys Into Uninfected Recipients: A Multi-Center Retrospective Cohort Study. Transpl Infect Dis 2025:e70011. [PMID: 40047349 DOI: 10.1111/tid.70011] [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: 11/27/2024] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Several studies have suggested an increased risk of cytomegalovirus (CMV) viremia among Hepatitis C virus (HCV)-uninfected recipients of kidney transplants from HCV-RNA+ deceased donors (HCV D+/R-), but these studies featured small sample sizes and limited ability to address confounding variables. METHODS We assembled a retrospective cohort of adult kidney transplant recipients at five US centers between 4/1/2015 and 12/31/2020 to determine the association between HCV D+/R- transplants and the outcomes of CMV viremia (> 1000 IU/mL), death-censored graft failure, and mortality in the first posttransplant year compared to HCV D-/R- transplants. We generated highly similar matched cohorts of HCV D+/R- and HCV D-/R- recipients based on attributes that affect the risk of CMV viremia. We matched exactly on center, CMV donor/recipient serostatus, and antibody induction therapy. RESULTS The cohort comprised 275 HCV D+/R- recipients with a mean age of 52.5 years (SD = 10.7); 19% were CMV D+/R-, and 74% received anti-thymocyte globulin induction. With variable ratio matching, 267 HCV D+/R- recipients were matched to 996 HCV D-/R- recipients. CMV viremia occurred in 15% of HCV D+/R- and 11% of HCV D-R- recipients. In Cox regression, transplantation with an HCV-RNA+ donor kidney was not associated with a significantly higher risk of CMV viremia (HR 1.3, 95% CI 0.89-1.92) or death-censored graft loss (HR 0.61, 95% CI 0.31-1.2). CONCLUSION The risk of CMV viremia was not significantly increased among HCV D+/R- kidney recipients. Future studies should examine associations between donor-derived HCV infection and clinical outcomes of CMV syndrome and disease.
Collapse
Affiliation(s)
- Vishnu S Potluri
- Renal-Electrolyte and Hypertension Division, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Goldberg
- Division of Digestive Health and Liver Disease, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Siqi Zhang
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas E Schaubel
- Department of Biostatistics, Epidemiology and Bioinformatics, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Miklos Z Molnar
- Department of Internal Medicine, Division of Nephrology & Hypertension, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah, USA
| | - Rachel Forbes
- Department of Surgery, Division of Kidney and Pancreas Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Megan E Sise
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - James L Rogers
- Department of Surgery, Division of Kidney and Pancreas Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Vasanthi Balaraman
- Division of Transplant Surgery, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Anshul Bhalla
- Department of Nephrology, Kaiser Permanente, San Francisco, California, USA
| | - David Shaffer
- Department of Surgery, Division of Kidney and Pancreas Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Beatrice P Concepcion
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Raymond T Chung
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ian A Strohbehn
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shristi Mapchan
- Renal-Electrolyte and Hypertension Division, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Akhila Sangadi
- Division of Digestive Health and Liver Disease, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Eric Martin
- Division of Digestive Health and Liver Disease, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Roy D Bloom
- Renal-Electrolyte and Hypertension Division, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alyssa Ammazzalorso
- Division of Infectious Disease, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emily A Blumberg
- Division of Infectious Disease, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter P Reese
- Department of Surgery, Division of Kidney and Pancreas Transplantation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Transplant Science, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
9
|
Sayadi H, Fromage Y, Labriffe M, Billat PA, Codde C, Arraki Zava S, Marquet P, Woillard JB. Estimation of Ganciclovir Exposure in Adults Transplant Patients by Machine Learning. AAPS J 2025; 27:53. [PMID: 40021573 DOI: 10.1208/s12248-025-01034-9] [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: 08/13/2024] [Accepted: 01/31/2025] [Indexed: 03/03/2025] Open
Abstract
INTRODUCTION Valganciclovir, a prodrug of ganciclovir (GCV), is used to prevent cytomegalovirus infection after transplantation, with doses adjusted based on creatinine clearance (CrCL) to target GCV AUC0-24 h of 40-60 mg*h/L. This sometimes leads to overexposure or underexposure. This study aimed to train, test and validate machine learning (ML) algorithms for accurate GCV AUC0-24 h estimation in solid organ transplantation. METHODS We simulated patients for different dosing regimen (900 mg/24 h, 450 mg/24 h, 450 mg/48 h, 450 mg/72 h) using two literature population pharmacokinetic models, allocating 75% for training and 25% for testing. Simulations from two other literature models and real patients provided validation datasets. Three independent sets of ML algorithms were created for each regimen, incorporating CrCL and 2 or 3 concentrations. We evaluated their performance on testing and validation datasets and compared them with MAP-BE. RESULTS XGBoost using 3 concentrations generated the most accurate predictions. In testing dataset, they exhibited a relative bias of -0.02 to 1.5% and a relative RMSE of 2.6 to 8.5%. In the validation dataset, a relative bias of 1.5 to 5.8% and 8.9 to 16.5%, and a relative RMSE of 8.5 to 9.6% and 10.7% to 19.7% were observed depending on the model used. XGBoost algorithms outperformed or matched MAP-BE, showing enhanced generalization and robustness in their estimates. When applied to real patients' data, algorithms using 2 concentrations showed relative bias of 1.26% and relative RMSE of 12.68%. CONCLUSIONS XGBoost ML models accurately estimated GCV AUC0-24 h from limited samples and CrCL, providing a strategy for optimized therapeutic drug monitoring.
Collapse
Affiliation(s)
- Hamza Sayadi
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
| | - Yeleen Fromage
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
| | - Marc Labriffe
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France
| | - Pierre-André Billat
- INERIS, Experimental Toxicology and Modeling Unit (TEAM), Parc ALATA BP2, Verneuil en Halatte, France
| | - Cyrielle Codde
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France
- Service de Maladies Infectieuses et Tropicales, CHU Dupuytren, Limoges, France
| | - Selim Arraki Zava
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
| | - Pierre Marquet
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France
| | - Jean-Baptiste Woillard
- Department of Pharmacology, Toxicology and Pharmacovigilance, CHU de Limoges, Limoges, France.
- Pharmacology & Transplantation, INSERM U1248, Université de Limoges, 2 Rue du Pr Descottes, F-87000, Limoges, France.
| |
Collapse
|
10
|
Arvanitis P, Davis MR, Farmakiotis D. Cytomegalovirus infection and cardiovascular outcomes in abdominal organ transplant recipients: A systematic review and meta-analysis. Transplant Rev (Orlando) 2024; 38:100860. [PMID: 38815340 PMCID: PMC11586461 DOI: 10.1016/j.trre.2024.100860] [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: 02/21/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Despite advancements in Cytomegalovirus (CMV) management, its impact on graft function, mortality, and cardiovascular (CV) health of organ transplant recipients (OTR) remains a significant concern. We investigated the association between CMV infection and CV events (CVE) in organ (other than heart) transplant recipients. METHODS We conducted a comprehensive literature search in PubMed and EMBASE, including studies that reported on CMV infection or disease and post-transplantation CVE. Studies of heart transplant recipients were excluded. RESULTS We screened 3875 abstracts and 12 clinical studies were included in the final analysis, mainly in kidney and liver transplant recipients. A significant association was observed between CMV infection and an increased risk of CVE, with a pooled unadjusted hazard ratio (HR) of 1.99 (95% Confidence Intervals [CI] 1.45-2.73) for CMV infection and 1.59 (95% CI 1.21-2.10) for CMV disease. Pooled adjusted HR were 2.17 (95% CI 1.47-3.20) and 1.77 (95% CI 0.83-3.76), respectively. Heterogeneity was low (I2 = 0%) for CMV infection, suggesting consistent association across studies, and moderate-to-high for CMVdisease (I2 = 50% for unadjusted, 53% for adjusted HR). DISCUSSION We found a significant association between CMV infection and CV risk in abdominal OTR, underscoring the importance of proactive CMV surveillance and early treatment. Future research should aim for more standardized methodologies to fully elucidate the relationship between CMV and CV outcomes, potentially informing novel preventive and therapeutic strategies that could benefit the CV health of OTR.
Collapse
Affiliation(s)
- Panos Arvanitis
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States
| | - Michel R Davis
- The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States
| | - Dimitrios Farmakiotis
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States.
| |
Collapse
|
11
|
Tilloy V, Díaz-González D, Laplace L, Bisserier E, Chou S, Rawlinson WD, Boivin G, Baldanti F, Lazzarotto T, Andrei G, Hirsch HH, Marcos MÁ, Michel D, Hantz S, Alain S. Comprehensive Herpesviruses Antiviral drug Resistance Mutation Database (CHARMD). Antiviral Res 2024; 231:106016. [PMID: 39349222 DOI: 10.1016/j.antiviral.2024.106016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
A comprehensive and accessible Herpesvirus drug resistance database was designed to serve as an international reference for diagnosis and clinical studies. This database available at https://www.unilim.fr/cnr-herpesvirus/outils/codexmv/includes both resistance-related mutations and natural polymorphisms. Initially designed for human cytomegalovirus, it will be expanded to include herpes simplex and varicella-zoster viruses. Newly published mutations and new mutations reported by users or collaborating expert laboratories will be reviewed by an international committee of reference laboratories before inclusion in the database. Coupled with the Herpesvirus Sequence Analysis tool (HSA) mutation reports from NGS or Sanger sequences, it will be an open source for researchers in the field of Herpesviruses. We hope to fill this unmet need for the development and standardization of resistance genotyping.
Collapse
Affiliation(s)
- Valentin Tilloy
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; UF9481 Bioinformatique, CHU Limoges, Limoges, France; UF8843 Génomique médicale, CHU Limoges, Limoges, France; Inserm, U1092, RESINFIT, Université de Limoges, Limoges, France.
| | - Daniel Díaz-González
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; UF8843 Génomique médicale, CHU Limoges, Limoges, France
| | - Lisa Laplace
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France
| | - Emilien Bisserier
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France
| | - Sunwen Chou
- Department of Veterans Affairs Medical Center, Portland. Oregon, USA
| | - William D Rawlinson
- Virology and OTDS Laboratories (SAViD), Prince of Wales Hospital, Barker Street, Randwick, NSW, 2031, NSW Health Pathology Randwick, Australia
| | - Guy Boivin
- Centre de recherche en infectiologie, CHU de Québec-Université Laval, Canada
| | | | - Tiziana Lazzarotto
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Graciela Andrei
- Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hans H Hirsch
- Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - María Ángeles Marcos
- Servicio de Microbiología, Hospital Clinic, ISGlobal (Instituto de Salud Global de Barcelona), Barcelona, Spain
| | | | - Sébastien Hantz
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; Inserm, U1092, RESINFIT, Université de Limoges, Limoges, France
| | - Sophie Alain
- Centre National de Référence des Herpèsvirus, CHU Limoges, Limoges, France; UF8843 Génomique médicale, CHU Limoges, Limoges, France; Inserm, U1092, RESINFIT, Université de Limoges, Limoges, France; FHU SUPORT, CHU Limoges, Limoges, France.
| |
Collapse
|
12
|
Yilmaz ZB, Memisoglu F, Akbulut S. Management of cytomegalovirus infection after liver transplantation. World J Transplant 2024; 14:93209. [PMID: 39295968 PMCID: PMC11317856 DOI: 10.5500/wjt.v14.i3.93209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/05/2024] [Accepted: 05/27/2024] [Indexed: 07/31/2024] Open
Abstract
Cytomegalovirus (CMV) infection is one of the primary causes of morbidity and mortality following liver transplantation (LT). Based on current worldwide guidelines, the most effective strategies for avoiding post-transplant CMV infection are antiviral prophylaxis and pre-emptive treatment. CMV- IgG serology is the established technique for pretransplant screening of both donors and recipients. The clinical presentation of CMV infection and disease exhibits variability, prompting clinicians to consistently consider this possibility, particularly within the first year post-transplantation or subsequent to heightened immunosuppression. At annual symposia to discuss CMV prevention and how treatment outcomes can be improved, evidence on the incorporation of immune functional tests into clinical practice is presented, and the results of studies with new antiviral treatments are evaluated. Although there are ongoing studies on the use of letermovir and maribavir in solid organ transplantation, a consensus reflected in the guidelines has not been formed. Determining the most appropriate strategy at the individual level appears to be the key to enhancing outcomes. Although prevention strategies reduce the risk of CMV disease, the disease can still occur in up to 50% of high-risk patients. A balance between the risk of infection and disease development and the use of immunosuppressants must be considered when talking about the proper management of CMV in solid organ transplant recipients. The objective of this study was to establish a comprehensive framework for the management of CMV in patients who have had LT.
Collapse
Affiliation(s)
- Zeynep Burcin Yilmaz
- Infectious Diseases and Clinical Microbiology, Inonu University Faculty of Medicine, Malatya 44280, Türkiye
| | - Funda Memisoglu
- Infectious Diseases and Clinical Microbiology, Inonu University Faculty of Medicine, Malatya 44280, Türkiye
| | - Sami Akbulut
- Surgery and Liver Transplant Institute, Inonu University Faculty of Medicine, Malatya 44280, Türkiye
| |
Collapse
|
13
|
Righi I, Barone I, Rosso L, Morlacchi LC, Rossetti V, Caffarena G, Limanaqi F, Palleschi A, Clerici M, Trabattoni D. Immunopathology of lung transplantation: from infection to rejection and vice versa. Front Immunol 2024; 15:1433469. [PMID: 39286256 PMCID: PMC11402714 DOI: 10.3389/fimmu.2024.1433469] [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: 05/15/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Lung transplantation offers a lifesaving option for patients with end-stage lung disease, but it is marred by a high risk of post-transplant infections, particularly involving multidrug-resistant bacteria, Cytomegalovirus, and fungal pathogens. This elevated infection rate, the highest among solid organ transplants, poses a significant challenge for clinicians, particularly within the first year post-transplantation, where infections are the leading cause of mortality. The direct exposure of lung allografts to the external environment exacerbates this vulnerability leading to constant immune stimulation and consequently to an elevated risk of triggering alloimmune responses to the lung allograft. The necessity of prolonged immunosuppression to prevent allograft rejection further complicates patient management by increasing susceptibility to infections and neoplasms, and complicating the differentiation between rejection and infection, which require diametrically opposed management strategies. This review explores the intricate balance between preventing allograft rejection and managing the heightened infection risk in lung transplant recipients.
Collapse
Affiliation(s)
- Ilaria Righi
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ivan Barone
- Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Lorenzo Rosso
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Letizia Corinna Morlacchi
- Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Valeria Rossetti
- Respiratory Unit and Cystic Fibrosis Adult Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanni Caffarena
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Department of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Fiona Limanaqi
- Department of Biomedical and Clinical Sciences (DIBIC), University of Milan, Milan, Italy
| | - Alessandro Palleschi
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Fondazione Don C. Gnocchi IRCCS, Milan, Italy
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences (DIBIC), University of Milan, Milan, Italy
| |
Collapse
|
14
|
Schmoeckel M, Längin M, Reichart B, Abicht JM, Bender M, Denner J, Marckmann G, Brenner P, Wolf E, Hagl C. [Xenotransplantation of solid organs]. CHIRURGIE (HEIDELBERG, GERMANY) 2024; 95:603-609. [PMID: 38748210 PMCID: PMC11286678 DOI: 10.1007/s00104-024-02093-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 07/30/2024]
Abstract
Transplantation of genetically modified porcine hearts and kidneys could become a solution to the persistent shortage of human organ donors. Progress has been made in genetic engineering of donor pigs, preservation techniques after organ harvesting and immunosuppression using co-stimulation blockade with anti-CD40/CD40L monoclonal antibodies. Progress has also been made in in the development of methods that detect pathogenic porcine viruses and prevent their transmission to the recipient. As normal land breed pig organs continue to grow in the recipient to their original size, different pig breeds (such as Auckland Island pigs) are now used which reach a final size suitable for humans. Alternatively, a knock-out of the growth hormone receptor gene has been established, e.g., in the 10GM genetically modified pigs from Revivicor/United Therapeutics, USA. The first clinical pilot studies including patients suffering from terminal heart failure are expected to start in Germany in about 2 years.
Collapse
Affiliation(s)
- Michael Schmoeckel
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum - Standort Großhadern, Marchioninistr. 15, 81377, München, Deutschland.
| | - Matthias Längin
- Klinik für Anästhesiologie, LMU Klinikum Großhadern, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Bruno Reichart
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, München, Deutschland
| | - Jan-Michael Abicht
- Klinik für Anästhesiologie, LMU Klinikum Großhadern, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Martin Bender
- Klinik für Anästhesiologie, LMU Klinikum Großhadern, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Joachim Denner
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Institut für Virologie, Fachbereich für Veterinärmedizin, FU Berlin, Berlin, Deutschland
| | - Georg Marckmann
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Institut für Ethik, Geschichte und Theorie der Medizin, LMU München, München, Deutschland
| | - Paolo Brenner
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum - Standort Großhadern, Marchioninistr. 15, 81377, München, Deutschland
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
| | - Eckhard Wolf
- DFG-Sonderforschungsbereich TR127 - Xenotransplantation, LMU München, München, Deutschland
- Genzentrum und Center for Innovative Medical Models (CIMM), LMU München, München, Deutschland
| | - Christian Hagl
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum - Standort Großhadern, Marchioninistr. 15, 81377, München, Deutschland
- Partner Site München, Deutsches Zentrum für Herz- und Kreislaufforschung e. V. (DZHK), München, Deutschland
| |
Collapse
|
15
|
Schmoeckel M, Längin M, Reichart B, Abicht JM, Bender M, Michel S, Kamla CE, Denner J, Tönjes RR, Schwinzer R, Marckmann G, Wolf E, Brenner P, Hagl C. Current Status of Cardiac Xenotransplantation: Report of a Workshop of the German Heart Transplant Centers, Martinsried, March 3, 2023. Thorac Cardiovasc Surg 2024; 72:273-284. [PMID: 38154473 PMCID: PMC11147670 DOI: 10.1055/a-2235-8854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/22/2023] [Indexed: 12/30/2023]
Abstract
This report comprises the contents of the presentations and following discussions of a workshop of the German Heart Transplant Centers in Martinsried, Germany on cardiac xenotransplantation. The production and current availability of genetically modified donor pigs, preservation techniques during organ harvesting, and immunosuppressive regimens in the recipient are described. Selection criteria for suitable patients and possible solutions to the problem of overgrowth of the xenotransplant are discussed. Obviously microbiological safety for the recipient and close contacts is essential, and ethical considerations to gain public acceptance for clinical applications are addressed. The first clinical trial will be regulated and supervised by the Paul-Ehrlich-Institute as the National Competent Authority for Germany, and the German Heart Transplant Centers agreed to cooperatively select the first patients for cardiac xenotransplantation.
Collapse
Affiliation(s)
- Michael Schmoeckel
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
| | - Matthias Längin
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Bruno Reichart
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Jan-Michael Abicht
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Martin Bender
- Klinik für Anaesthesiologie, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Sebastian Michel
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | | | - Joachim Denner
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Institut für Virologie, Fachbereich für Veterinärmedizin, Freie Universität Berlin, Berlin, Germany
| | - Ralf Reinhard Tönjes
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Paul-Ehrlich-Institut, Langen, Germany
| | - Reinhard Schwinzer
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Georg Marckmann
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Institut für Ethik, Geschichte und Theorie der Medizin, LMU München, Germany
| | - Eckhard Wolf
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
- Lehrstuhl für Molekulare Tierzucht und Biotechnologie, Genzentrum der LMU München, Germany
| | - Paolo Brenner
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
- DFG-Transregio-Sonderforschungsbereich TR127—Xenotransplantation, Walter-Brendel-Zentrum für Experimentelle Medizin, LMU München, Germany
| | - Christian Hagl
- Herzchirurgische Klinik und Poliklinik, LMU Klinikum, LMU München, Germany
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), Partner Site Munich, Germany
| |
Collapse
|
16
|
Schneider M, Kollender K, Hilfrich B, Weiss R, Iftner T, Heim A, Ganzenmueller T. Evaluation of an automated real-time transcription-mediated amplification (TMA) assay for detection and quantification of cytomegalovirus DNA in different clinical specimens. J Clin Virol 2024; 171:105637. [PMID: 38218116 DOI: 10.1016/j.jcv.2023.105637] [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: 10/26/2023] [Revised: 12/13/2023] [Accepted: 12/31/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Reliable and fast detection and quantification of human cytomegalovirus (CMV) DNA in various diagnostic specimens is essential for care of immunocompromised or congenitally infected individuals. OBJECTIVES To evaluate the analytical and clinical performance of the Panther Aptima® CMV (Hologic) quantitative real-time transcription mediated amplification (TMA) assay. STUDY DESIGN Performance of the TMA assay run on the Hologic Panther Fusion was analysed for 32 proficiency testing samples and 21 quantitative reproducibility panel samples; additionally, we compared results of TMA assay and routine quantitative real-time PCR assays ("PCR-A"= Biomérieux CMV R-gene® or "PCR-B"= Laboratory-developed CMV-PCR) in 518 diagnostic specimens (254 plasma, 120 EDTA whole blood, 43 urine, 45 amniotic fluid and 56 breast milk) at two university hospital laboratories. RESULTS All proficiency panel samples were correctly identified and quantified by the TMA assay; replicate testing of the reproducibility panel samples showed good reproducibility within and between the two laboratories. Sensitivity in plasma and WB was higher for the TMA assay detecting low-level CMV-DNAemia in samples tested negative by routine PCR. Quantitative CMV-DNAemia values correlated well between TMA and real-time PCR. Similarly, urine, AF and BM specimens showed a high rate of concordant results (91%, 98% and 98%, respectively) among TMA and PCR with good correlation of quantitative values. CONCLUSION The performance of the Aptima® CMV TMA assay for viral blood load testing compared well to established real-time PCRs. In addition, it can be useful for diagnostics in urine, amniotic fluid and breast milk specimens.
Collapse
Affiliation(s)
- M Schneider
- University Hospital Tuebingen, Institute for Medical Virology and Epidemiology of Viral Diseases, Tuebingen, Germany
| | - K Kollender
- University Hospital Tuebingen, Institute for Medical Virology and Epidemiology of Viral Diseases, Tuebingen, Germany; University Hospital Tuebingen, Consiliary Laboratory for congenital and postnatal CMV infections, Tuebingen, Germany
| | - B Hilfrich
- Hannover Medical School, Institute of Virology, Hannover, Germany
| | - R Weiss
- University Hospital Tuebingen, Institute for Medical Virology and Epidemiology of Viral Diseases, Tuebingen, Germany
| | - T Iftner
- University Hospital Tuebingen, Institute for Medical Virology and Epidemiology of Viral Diseases, Tuebingen, Germany; German Center for Infection Research (DZIF), Site Tuebingen, Tuebingen, Germany
| | - A Heim
- Hannover Medical School, Institute of Virology, Hannover, Germany
| | - T Ganzenmueller
- University Hospital Tuebingen, Institute for Medical Virology and Epidemiology of Viral Diseases, Tuebingen, Germany; University Hospital Tuebingen, Consiliary Laboratory for congenital and postnatal CMV infections, Tuebingen, Germany; German Center for Infection Research (DZIF), Site Tuebingen, Tuebingen, Germany.
| |
Collapse
|
17
|
Huelsboemer L, Boroumand S, Kochen A, Dony A, Moscarelli J, Hauc SC, Stögner VA, Formica RN, Pomahac B, Kauke-Navarro M. Immunosuppressive strategies in face and hand transplantation: a comprehensive systematic review of current therapy regimens and outcomes. FRONTIERS IN TRANSPLANTATION 2024; 3:1366243. [PMID: 38993787 PMCID: PMC11235358 DOI: 10.3389/frtra.2024.1366243] [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: 01/05/2024] [Accepted: 02/21/2024] [Indexed: 07/13/2024]
Abstract
Background Recipients of Vascularized Composite Allotransplants require effective immunosuppressive therapy to prevent graft rejection. This systematic review summarizes the current body of literature on immunosuppressive regimens used in face and hand transplants while summarizing their outcome in terms of rejection, renal failure, and infections. Methods A systematic search of electronic databases was conducted to identify relevant studies from 1998 until July 1st, 2023. We included all studies that discussed immunosuppressive strategies in face and hand transplant recipients according to PRISMA. Results The standard triple maintenance therapy was mostly adjusted due to nephrotoxicity or high incidence of rejection. The most common alternative treatments utilized were sirolimus (25/91; 27.5%) or everolimus (9/91; 9.9%) following hand- and photophoresis (7/45; 15.6%), sirolimus (5/45; 11.1%) or belatacept (1/45; 2.2%) following face transplantation. Episodes of rejection were reported in 60 (65.9%) of hand- and 33 (73%) of face transplant patients respectively. Graft loss of 12 (13.2%) hand and 4 (8.9%) face transplants was reported. Clinical CMV infection was observed in 6 (6.6%) hand and 7 (15.5%) face transplant recipients. Conclusions Based on the herein presented data, facial grafts exhibited a heightened incidence of rejection episodes and CMV infections. Facial mucosa adds complexity to the immunological graft composition highlighting the need of individualized immunosuppressive regimens and further research.
Collapse
Affiliation(s)
- Lioba Huelsboemer
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Sam Boroumand
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Alejandro Kochen
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
- Regenerative Wound Healing Center, Yale School of Medicine, New Haven, CT, United States
| | - Alna Dony
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Jake Moscarelli
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Sacha C. Hauc
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Viola A. Stögner
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Richard N. Formica
- Department of Medicine, Section of Nephrology and Transplantation, Yale School of Medicine, New Haven, CT, United States
| | - Bohdan Pomahac
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Martin Kauke-Navarro
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
18
|
Otto WR, Vora SB, Dulek DE. Cytomegalovirus Cell-mediated Immunity Assays in Pediatric Transplantation. J Pediatric Infect Dis Soc 2024; 13:S22-S30. [PMID: 38417088 DOI: 10.1093/jpids/piae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/23/2024] [Indexed: 03/01/2024]
Abstract
Cytomegalovirus (CMV) is a significant cause of morbidity and mortality in pediatric transplantation. However, currently utilized CMV prevention paradigms have limitations, leading to research aimed at novel strategies for mitigation of CMV infection. Cell-mediated immunity (CMI) is crucial in controlling CMV infection and the use of CMV-specific CMI assays to guide prevention and treatment of CMV infection in both solid organ transplant and hematopoietic cell transplant recipients shows great promise. In this article, we review the immune response to CMV infection to highlight the rationale for CMI assays, describe available commercial assays and strategies for their use, and summarize relevant literature regarding the use of CMI assays in transplant recipients.
Collapse
Affiliation(s)
- William R Otto
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Surabhi B Vora
- Division of Infectious Diseases, Seattle Children's Hospital, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Daniel E Dulek
- Division of Pediatric Infectious Diseases, Monroe Carell Jr Children's Hospital at Vanderbilt, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
19
|
Rocha FA, Silveira CRF, Dos Santos AF, Stefanini ACB, Hamerschlak N, Marti LC. Development of a highly cytotoxic, clinical-grade virus-specific T cell product for adoptive T cell therapy. Cell Immunol 2024; 395-396:104795. [PMID: 38101075 DOI: 10.1016/j.cellimm.2023.104795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023]
Abstract
At present, recipients of allogeneic hematopoietic stem-cells are still suffering from recurrent infections after transplantation. Infusion of virus-specific T cells (VST) post-transplant reportedly fights several viruses without increasing the risk of de novo graft-versus-host disease. This study targeted cytomegalovirus (CMV) for the development of an innovative approach for generating a very specific VST product following Good Manufacturing Practices (GMP) guidelines. We used a sterile disposable compartment named the Leukoreduction System Chamber (LRS-chamber) from the apheresis platelet donation kit as the starting material, which has demonstrated high levels of T cells. Using a combination of IL-2 and IL-7 we could improve expansion of CMV-specific T cells. Moreover, by developing and establishing a new product protocol, we were able to stimulate VST proliferation and favors T cell effector memory profile. The expanded VST were enriched in a closed automated system, creating a highly pure anti-CMV product, which was pre-clinically tested for specificity in vitro and for persistence, biodistribution, and toxicity in vivo using NOD scid mice. Our results demonstrated very specific VST, able to secrete high amounts of interferon only in the presence of cells infected by the human CMV strain (AD169), and innocuous to cells partially HLA compatible without viral infection.
Collapse
Affiliation(s)
- Fernanda Agostini Rocha
- Hospital Israelita Albert Einstein, Department of Experimental Research, Rua Comendador Elias Jafet, 755 Zip code: 05653 000, São Paulo, SP, Brazil
| | - Caio Raony Farina Silveira
- Hospital Israelita Albert Einstein, Department of Experimental Research, Rua Comendador Elias Jafet, 755 Zip code: 05653 000, São Paulo, SP, Brazil
| | - Ancély Ferreira Dos Santos
- Hospital Israelita Albert Einstein, Department of Experimental Research, Rua Comendador Elias Jafet, 755 Zip code: 05653 000, São Paulo, SP, Brazil
| | - Ana Carolina Buzzo Stefanini
- Hospital Israelita Albert Einstein, Department of Experimental Research, Rua Comendador Elias Jafet, 755 Zip code: 05653 000, São Paulo, SP, Brazil
| | - Nelson Hamerschlak
- Hospital Israelita Albert Einstein, Department of Bone Marrow Transplant, Avenida Albert Einstein, 627 Zip code: 05652 000, São Paulo, SP, Brazil
| | - Luciana Cavalheiro Marti
- Hospital Israelita Albert Einstein, Department of Experimental Research, Rua Comendador Elias Jafet, 755 Zip code: 05653 000, São Paulo, SP, Brazil.
| |
Collapse
|
20
|
Malagola M, Radici V, Farina M, Pellizzeri S, Spoldi F, Morello E, Polverelli N, Buttini EA, Bernardi S, Re F, Leoni A, Signorini L, Caruso A, Russo D. CMV prophylaxis with letermovir significantly improves graft and relapse free survival following allogeneic stem cell transplantation. Bone Marrow Transplant 2024; 59:138-140. [PMID: 37857841 PMCID: PMC10781629 DOI: 10.1038/s41409-023-02124-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Affiliation(s)
- Michele Malagola
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
| | - Vera Radici
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Mirko Farina
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simone Pellizzeri
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Filippo Spoldi
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Enrico Morello
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Nicola Polverelli
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eugenia Accorsi Buttini
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simona Bernardi
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA), "ASST-Spedali Civili" Hospital, Brescia, Italy
| | - Federica Re
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA), "ASST-Spedali Civili" Hospital, Brescia, Italy
| | - Alessandro Leoni
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Centro di Ricerca Emato-oncologico AIL (CREA), "ASST-Spedali Civili" Hospital, Brescia, Italy
| | - Liana Signorini
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Arnaldo Caruso
- Institute of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, ASST Spedali Civili, Brescia, Italy
| | - Domenico Russo
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, "ASST-Spedali Civili" Hospital of Brescia; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
21
|
von Hoerschelmann E, Münch J, Gao L, Lücht C, Naik MG, Schmidt D, Pitzinger P, Michel D, Avaniadi P, Schrezenmeier E, Choi M, Halleck F, Budde K. Letermovir Rescue Therapy in Kidney Transplant Recipients with Refractory/Resistant CMV Disease. J Clin Med 2023; 13:100. [PMID: 38202107 PMCID: PMC10780128 DOI: 10.3390/jcm13010100] [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: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
(1) Background: CMV infections remain a problem after kidney transplantation, particularly if patients are refractory or resistant (r/r) to treatment with valganciclovir (VGCV) or ganciclovir (GCV). (2) Methods: In a single-center retrospective study, kidney transplant recipients (KTR) receiving letermovir (LTV) as rescue therapy for VGCV-/GCV-r/r CMV disease were analyzed regarding CMV history, immunosuppression, and outcomes. (3) Results: Of 201 KTR treated for CMV between 2017 and 2022, 8 patients received LTV following treatment failure with VGCV/GCV. All patients received CMV prophylaxis with VGCV according to the center's protocol, and 7/8 patients had a high-risk (D+/R-) CMV constellation. In seven of eight cases, rising CMV levels occurred during prophylaxis. In seven of eight patients, a mutation in UL97 associated with a decreased response to VGCV/GCV was detected. In four of eight patients, LTV resulted in CMV clearance after 24 ± 10 weeks (16-39 weeks), two of eight patients stabilized at viral loads <2000 cop/mL (6-20 weeks), and two of eight patients developed LTV resistance (range 8-10 weeks). (4) Conclusion: LTV, which is currently evaluated for CMV prophylaxis in kidney transplantation, also shows promising results for the treatment of patients with VGCV/GCV resistance despite the risk of developing LTV resistance. Additional studies are needed to further define its role in the treatment of patients with CMV resistance.
Collapse
Affiliation(s)
- Ellen von Hoerschelmann
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Johannes Münch
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Linde Gao
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Christian Lücht
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Marcel G. Naik
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Paul Pitzinger
- Institute of Virology, Charité Universitätsmedizin Berlin, Labor Berlin-Charité-Vivantes GmbH, 10117 Berlin, Germany
| | - Detlef Michel
- Institute of Virology, Universitätsklinikum Ulm, 89081 Ulm, Germany
| | - Parthenopi Avaniadi
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
22
|
Demirhan S, Munoz FM, Valencia Deray KG, Bocchini CE, Danziger-Isakov L, Blum S, Sharma TS, Sherman G, Boguniewicz J, Bacon S, Ardura MI, Maron GM, Ferrolino J, Foca M, Herold BC. Body surface area compared to body weight dosing of valganciclovir is associated with increased toxicity in pediatric solid organ transplantation recipients. Am J Transplant 2023; 23:1961-1971. [PMID: 37499799 DOI: 10.1016/j.ajt.2023.07.013] [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: 04/15/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Optimal dosing of valganciclovir (VGCV) for cytomegalovirus (CMV) prevention in pediatric solid organ transplantation recipients (SOTR) is controversial. Dosing calculated based on body surface area (BSA) and creatinine clearance is recommended but simplified body weight (BW) dosing is often prescribed. We conducted a retrospective 6-center study to compare safety and efficacy of these strategies in the first-year posttransplant There were 100 (24.2%) pediatric SOTR treated with BSA and 312 (75.7%) with BW dosing. CMV DNAemia was documented in 31.0% vs 23.4% (P = .1) at any time during the first year and breakthrough DNAemia in 16% vs 12.2% (P = .3) of pediatric SOTR receiving BSA vs BW dosing, respectively. However, neutropenia (50% vs 29.3%, P <.001), lymphopenia (51% vs 15.0%, P <.001), and acute kidney injury causing treatment modification (8.0% vs 1.8%, P <.001) were documented more frequently during prophylaxis in pediatric SOTR receiving BSA vs BW dosing. The adjusted odds ratio of VGCV-attributed toxicities comparing BSA and BW dosing was 2.3 (95% confidence interval [CI], 1.4-3.7] for neutropenia, 7.0 (95% CI, 3.9-12.4) for lymphopenia, and 4.6 (95% CI, 2.2-9.3) for premature discontinuation or dose reduction of VGCV, respectively. Results demonstrate that BW dosing is associated with significantly less toxicity without any increase in CMV DNAemia.
Collapse
Affiliation(s)
- Salih Demirhan
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Flor M Munoz
- Department of Pediatrics, Division of Infectious Diseases, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Kristen G Valencia Deray
- Department of Pediatrics, Division of Infectious Diseases, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Claire E Bocchini
- Department of Pediatrics, Division of Infectious Diseases, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Lara Danziger-Isakov
- Department of Pediatrics, Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Samantha Blum
- Department of Pediatrics, Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Tanvi S Sharma
- Department of Pediatrics, Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gilad Sherman
- Department of Pediatrics, Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Juri Boguniewicz
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Samantha Bacon
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Monica I Ardura
- Department of Pediatrics, Division of Infectious Diseases & Host Defense, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Gabriela M Maron
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jose Ferrolino
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Marc Foca
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA.
| | - Betsy C Herold
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
23
|
Bestard O, Kaminski H, Couzi L, Fernández-Ruiz M, Manuel O. Cytomegalovirus Cell-Mediated Immunity: Ready for Routine Use? Transpl Int 2023; 36:11963. [PMID: 38020746 PMCID: PMC10661902 DOI: 10.3389/ti.2023.11963] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
Utilizing assays that assess specific T-cell-mediated immunity against cytomegalovirus (CMV) holds the potential to enhance personalized strategies aimed at preventing and treating CMV in organ transplantation. This includes improved risk stratification during transplantation compared to relying solely on CMV serostatus, as well as determining the optimal duration of antiviral prophylaxis, deciding on antiviral therapy when asymptomatic replication occurs, and estimating the risk of recurrence. In this review, we initially provide an overlook of the current concepts into the immune control of CMV after transplantation. We then summarize the existent literature on the clinical experience of the use of immune monitoring in organ transplantation, with a particular interest on the outcomes of interventional trials. Current evidence indicates that cell-mediated immune assays are helpful in identifying patients at low risk for replication for whom preventive measures against CMV can be safely withheld. As more data accumulates from these and other clinical scenarios, it is foreseeable that these assays will likely become part of the routine clinical practice in organ transplantation.
Collapse
Affiliation(s)
- Oriol Bestard
- Nephrology and Kidney Transplant Department, Vall Hebron University Hospital, Barcelona, Spain
- Nephrology and Kidney Transplant Research Laboratory, Vall Hebrón Institut de Recerca (VHIR), Barcelona, Spain
| | - Hannah Kaminski
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
- UMR 5164-ImmunoConcEpT, University of Bordeaux, Centre National de la Recherche Scientifique (CNRS), Bordeaux University, Bordeaux, France
| | - Lionel Couzi
- Department of Nephrology, Transplantation, Dialysis and Apheresis, Centre Hospitalier Universitaire Bordeaux, Bordeaux, France
- UMR 5164-ImmunoConcEpT, University of Bordeaux, Centre National de la Recherche Scientifique (CNRS), Bordeaux University, Bordeaux, France
| | - Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario “12 de Octubre”, Instituto de Investigación Sanitaria Hospital “12 de Octubre” (imas12), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Oriol Manuel
- Infectious Diseases Service and Transplantation Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
24
|
Leuzinger K, Hirsch HH. Amplicon size and non-encapsidated DNA fragments define plasma cytomegalovirus DNA loads by automated nucleic acid testing platforms: A marker of viral cytopathology? J Med Virol 2023; 95:e29139. [PMID: 37804497 DOI: 10.1002/jmv.29139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/31/2023] [Accepted: 09/20/2023] [Indexed: 10/09/2023]
Abstract
Management of cytomegalovirus (CMV) in transplant patients relies on measuring plasma CMV-loads using quantitative nucleic acid testing (QNAT). We prospectively compared the automated Roche-cobas®6800-CMV and Roche-CAP/CTM-CMV with laboratory-developed Basel-CMV-UL54-95bp, and Basel-CMV-UL111a-77bp. Roche-cobas®6800-CMV and Roche-CAP/CTM-CMV were qualitatively concordant in 142/150 cases (95%). In-depth comparison revealed higher CMV-loads of the laboratory-developed assay and correlated with smaller amplicon size. After calibration to the 1.WHO-approved CMV international standard, differences were reduced but remained significant. DNase-I pretreatment significantly reduced CMV-loads for both automated Roche-CAP/CTM-CMV and Roche-cobas®6800-CMV assays, whereby 90% and 95% of samples became undetectable. DNase-I pretreatment also reduced CMV-loads quantified by Basel-CMV-UL54-95bp and Basel-CMV-UL111a-77bp, but remaining detectable in 20% and 35%, respectively. Differences were largest for 110 samples with low-level CMV-DNAemia being detectable but not-quantifiable by Roche-cobas®6800-CMV, whereby the smaller amplicon sizes yielded higher viral loads for concordant positives. We conclude that non-encapsidated fragmented CMV-DNA is the major form of plasma CMV-loads also measured by fully-automated platforms. Amplicons of <150 bp and calibrators are needed for reliable and commutable QNAT-results. We hypothesize that non-encapsidated fragmented CMV-DNA results from lysis of CMV-replicating cells and represent a direct marker of viral cell damage, which contribute to delayed viral load responses despite effective antivirals.
Collapse
Affiliation(s)
- Karoline Leuzinger
- Clinical Virology, University Hospital Basel, Basel, Switzerland
- Transplantation & Clinical Virology, Department Biomedicine, University of Basel, Basel, Switzerland
| | - Hans H Hirsch
- Clinical Virology, University Hospital Basel, Basel, Switzerland
- Transplantation & Clinical Virology, Department Biomedicine, University of Basel, Basel, Switzerland
- Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
25
|
Zhang T, Potgieter TI, Kosche E, Rückert J, Ostermann E, Schulz T, Empting M, Brune W. Thioxothiazolo[3,4-a]quinazoline derivatives inhibit the human cytomegalovirus alkaline nuclease. Antiviral Res 2023; 217:105696. [PMID: 37541625 DOI: 10.1016/j.antiviral.2023.105696] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Human cytomegalovirus (HCMV, human herpesvirus 5) is an opportunistic pathogen responsible for serious disease in immunocompromised patients. Current antiviral therapies rely predominantly on drugs interfering with viral DNA replication and packaging. However, the serious side effects of existing drugs and the emergence of drug resistance indicate the need for new targets for anti-HCMV therapy. One such target is the viral alkaline nuclease (AN), an enzyme highly conserved among the Herpesviridae. In this study, we validated the HCMV AN, encoded by the viral UL98 open reading frame, as a drug target by demonstrating that a UL98-deficient HCMV mutant is severely attenuated and shows a reduced ability to spread in cell culture. We established a fluorescence-based enzyme assay suitable for high-throughput screening and used it on a small-molecule compound library. The most promising hit, a thioxothiazolo[3,4-a]quinazoline derivative, blocked AN activity in vitro and inhibited HCMV replication in plaque reduction (PRA) and fluorescence reduction assays (FRA). Several derivatives of the hit compound were tested, some of which had similar or better inhibitory activities. The most potent derivative of hit scaffold A, compound AD-51, inhibited HCMV replication with a 50% effective concentrations (EC50) of 0.9 μM in the FRA and 1.1 μM in the PRA. AD-51 was also active against ganciclovir, foscarnet, and letermovir-resistant HCMVs. Moreover, it inhibited herpes simplex virus, Kaposi's sarcoma-associated herpesvirus, and murine CMV, a mouse virus serving as a model for HCMV. These results suggest that thioxothiazolo[3,4-a]quinazoline derivatives are a new class of herpesvirus inhibitors targeting the viral AN.
Collapse
Affiliation(s)
- Tianyu Zhang
- Leibniz Institute of Virology (LIV), Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Theodore I Potgieter
- Leibniz Institute of Virology (LIV), Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Erik Kosche
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany; German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Jessica Rückert
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Thomas Schulz
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Martin Empting
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany; German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Germany
| | - Wolfram Brune
- Leibniz Institute of Virology (LIV), Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Germany.
| |
Collapse
|
26
|
Malahe SRK, van Kampen JJA, Manintveld OC, Hoek RAS, den Hoed CM, Baan CC, Kho MML, Verjans GMGM. Current Perspectives on the Management of Herpesvirus Infections in Solid Organ Transplant Recipients. Viruses 2023; 15:1595. [PMID: 37515280 PMCID: PMC10383436 DOI: 10.3390/v15071595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Solid organ transplant recipients (SOTRs) are at high risk of human herpesvirus (HHV)-related morbidity and mortality due to the use of immunosuppressive therapy. We aim to increase awareness and understanding of HHV disease burden in SOTRs by providing an overview of current prevention and management strategies as described in the literature and guidelines. We discuss challenges in both prevention and treatment as well as future perspectives.
Collapse
Affiliation(s)
- S Reshwan K Malahe
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jeroen J A van Kampen
- Department of Viroscience, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Olivier C Manintveld
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Cardiology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Rogier A S Hoek
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Caroline M den Hoed
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Marcia M L Kho
- Department of Internal Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Transplant Institute, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Georges M G M Verjans
- Department of Viroscience, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- HerpeslabNL, Department of Viroscience, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
27
|
Crawford LB. Hematopoietic stem cells and betaherpesvirus latency. Front Cell Infect Microbiol 2023; 13:1189805. [PMID: 37346032 PMCID: PMC10279960 DOI: 10.3389/fcimb.2023.1189805] [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: 03/20/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
The human betaherpesviruses including human cytomegalovirus (HCMV), human herpesvirus (HHV)-6a and HHV-6b, and HHV-7 infect and establish latency in CD34+ hematopoietic stem and progenitor cells (HPCs). The diverse repertoire of HPCs in humans and the complex interactions between these viruses and host HPCs regulate the viral lifecycle, including latency. Precise manipulation of host and viral factors contribute to preferential maintenance of the viral genome, increased host cell survival, and specific manipulation of the cellular environment including suppression of neighboring cells and immune control. The dynamic control of these processes by the virus regulate inter- and intra-host signals critical to the establishment of chronic infection. Regulation occurs through direct viral protein interactions and cellular signaling, miRNA regulation, and viral mimics of cellular receptors and ligands, all leading to control of cell proliferation, survival, and differentiation. Hematopoietic stem cells have unique biological properties and the tandem control of virus and host make this a unique environment for chronic herpesvirus infection in the bone marrow. This review highlights the elegant complexities of the betaherpesvirus latency and HPC virus-host interactions.
Collapse
Affiliation(s)
- Lindsey B Crawford
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
28
|
Fernández-Ruiz M, Redondo N, Parra P, Ruiz-Merlo T, Rodríguez-Goncer I, Polanco N, González E, López-Medrano F, San Juan R, Navarro D, Andrés A, Aguado JM. Comparison of intracellular cytokine staining versus an ELISA-based assay to assess CMV-specific cell-mediated immunity in high-risk kidney transplant recipients. J Med Virol 2023; 95:e28733. [PMID: 37185851 DOI: 10.1002/jmv.28733] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023]
Abstract
The best method for monitoring cytomegalovirus (CMV)-specific cell-mediated immunity (CMV-CMI) among high-risk kidney transplant (KT) recipients remains uncertain. We assessed CMV-CMI by intracellular cytokine staining (ICS) by flow cytometry and a commercial interferon (IFN)-γ release assay (QuantiFERON®-CMV [QTF-CMV]) at posttransplant months 3, 4, and 5 in 53 CMV-seropositive KT recipients that had received induction therapy with antithymocyte globulin (ATG) and a 3-month course of valganciclovir prophylaxis. The discriminative capacity (areas under receiver operating characteristics curve [auROCs]) and diagnostic accuracy to predict immune protection against CMV infection from the discontinuation of prophylaxis to month 12 were compared between both methods. There was significant although moderate correlations between CMV-specific IFN-γ-producing CD8+ T-cell counts enumerated by ICS and IFN-γ levels by QTF-CMV at months 3 (rho: 0.493; p = 0.005) and 4 (rho: 0.440; p = 0.077). The auROCs for CMV-specific CD4+ and CD8+ T-cells by ICS were nonsignificantly higher than that of QTF-CMV (0.696 and 0.733 vs. 0.678; p = 0.900 and 0.692, respectively). The optimal cut-off of ≥0.395 CMV-specific CD8+ T-cells yielded a sensitivity of 86.4%, specificity of 54.6%, positive predictive value of 79.2% and negative predictive value of 66.7% to predict protection. The corresponding estimates for QTF-CMV (IFN-γ levels ≥0.2 IU/mL) were 78.9%, 37.5%, 75.0%, and 42.9%, respectively. The enumeration of CMV-specific IFN-γ-producing CD8+ T-cells at the time of cessation of prophylaxis performed slightly better than the QTF-CMV assay to predict immune protection in seropositive KT recipients previously treated with ATG.
Collapse
Affiliation(s)
- Mario Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Natalia Redondo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Patricia Parra
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Tamara Ruiz-Merlo
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Isabel Rodríguez-Goncer
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Natalia Polanco
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Esther González
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - Francisco López-Medrano
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Rafael San Juan
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| | - David Navarro
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Microbiology, Hospital Clínico Universitario, Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Amado Andrés
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
- Department of Nephrology, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
| | - José María Aguado
- Unit of Infectious Diseases, Hospital Universitario "12 de Octubre", Instituto de Investigación Sanitaria Hospital "12 de Octubre" (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense, Madrid, Spain
| |
Collapse
|