1
|
Swan CL, Dushimiyimana V, Ndishimye P, Buchanan R, Yourkowski A, Semafara S, Nsanzimana S, Francis ME, Thivierge B, Lew J, Facciuolo A, Gerdts V, Falzarano D, Sjaarda C, Kelvin DJ, Bitunguhari L, Kelvin AA. Third COVID-19 vaccine dose boosts antibody function in Rwandans with high HIV viral load. iScience 2023; 26:107959. [PMID: 37810226 PMCID: PMC10558770 DOI: 10.1016/j.isci.2023.107959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/18/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023] Open
Abstract
SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) causing COVID-19 (coronavirus disease 2019) poses a greater health risk to immunocompromized individuals including people living with HIV (PLWH). However, most studies on PLWH have been conducted in higher-income countries. We investigated the post-vaccination antibody responses of PLWH in Rwanda by collecting peripheral blood from participants after receiving a second or third COVID-19 vaccine. Virus-binding antibodies as well as antibody neutralization ability against all major SARS-CoV-2 variants of concern were analyzed. We found that people with high HIV viral loads and two COVID-19 vaccine doses had lower levels of binding antibodies that were less virus neutralizing and less cross-reactive compared to control groups. A third vaccination increased neutralizing antibody titers. Our data suggest that people with high HIV viral loads require a third dose of vaccine to neutralize SARS-CoV-2 virus and new variants as they emerge.
Collapse
Affiliation(s)
- Cynthia L. Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | | | - Pacifique Ndishimye
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- African Institute for Mathematical Sciences, Kigali, Rwanda
| | - Rachelle Buchanan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Sage Semafara
- Rwanda Network of the People living with HIV (RRP+), Kigali, Rwanda
| | | | - Magen E. Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Brittany Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Jocelyne Lew
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Antonio Facciuolo
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Calvin Sjaarda
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
- Queen’s Genomics Lab at Ongwanada (Q-GLO), Ongwanada Resource Centre, Kingston, ON K7M 8A6, Canada
| | - David J. Kelvin
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
2
|
von Moos S, Rho E, Dammann M, Kokkonen SM, Mueller TF, Schachtner T. Therapeutic Drug Monitoring of Mycophenolic Acid Identifies Kidney Transplant Recipients Responsive to Two SARS-CoV-2 mRNA Vaccine Doses. Transpl Int 2023; 36:11286. [PMID: 37448450 PMCID: PMC10336200 DOI: 10.3389/ti.2023.11286] [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: 02/18/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023]
Abstract
Immune-responsiveness to SARS-CoV-2 mRNA vaccination is reduced in kidney transplant recipients (KTRs). Previous reports point to a role of mycophenolic acid (MPA). Our observational cohort study included all KTRs at University Hospital Zurich receiving two SARS-CoV-2 mRNA vaccine doses more than 6 months post-transplantation, who were assessed by measuring anti-spike immunoglobulin G (IgG). We applied principles of therapeutic drug monitoring (TDM) to correlate MPA exposure and lymphocyte counts with SARS-CoV-2 IgG. MPA trough levels differ largely among KTRs with a median of 3.1 mg/L (range 0.7-9.5 mg/L). 34 of 84 KTRs (40%) developed positive SARS-CoV-2 IgG after two vaccine doses. KTRs who developed positive SARS-CoV-2 IgG showed significantly higher eGFR (p < 0.001), lower MPA trough levels (p < 0.001) and higher CD19+ lymphocytes (p < 0.001). MPA trough levels <2.5 mg/l and CD19+ lymphocytes >40/μl identify KTRs with seroconversion. Upon logistic regression, MPA trough levels <2.5 mg/L were associated with a 7-fold (CI 95%: 1.589-29.934) and ciclosporin use with a 6-fold (CI 95%: 1.148-30.853) increase in the odds of seroconversion. Our study indicates that immune-responsiveness to SARS-CoV-2 mRNA vaccines correlates with MPA exposure measured by MPA trough level but argues against a class effect of MPA. TDM-guided MPA dosing may be a strategy to increase seroconversion rate.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas Schachtner
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Banjongjit A, Phirom S, Phannajit J, Jantarabenjakul W, Paitoonpong L, Kittanamongkolchai W, Wattanatorn S, Prasithsirikul W, Eiam-Ong S, Avihingsanon Y, Hansasuta P, Vanichanan J, Townamchai N. Benefits of Switching Mycophenolic Acid to Sirolimus on Serological Response after a SARS-CoV-2 Booster Dose among Kidney Transplant Recipients: A Pilot Study. Vaccines (Basel) 2022; 10:vaccines10101685. [PMID: 36298550 PMCID: PMC9609831 DOI: 10.3390/vaccines10101685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022] Open
Abstract
Kidney transplant recipients (KTRs) have a suboptimal immune response to COVID-19 vaccination due to the effects of immunosuppression, mostly mycophenolic acid (MPA). This study investigated the benefits of switching from the standard immunosuppressive regimen (tacrolimus (TAC), MPA, and prednisolone) to a regimen of mammalian target of rapamycin inhibitor (mTORi), TAC and prednisolone two weeks pre- and two weeks post-BNT162b2 booster vaccination. A single-center, opened-label pilot study was conducted in KTRs, who received two doses of ChAdOx-1 and a single dose of BNT162b2. The participants were randomly assigned to continue the standard regimen (control group, n = 14) or switched to a sirolimus (an mTORi), TAC, and prednisolone (switching group, n = 14) regimen two weeks before and two weeks after receiving a booster dose of BNT162b2. The anti-SARS-CoV-2 S antibody level after vaccination in the switching group was significantly greater than the control group (4051.0 [IQR 3142.0-6466.0] BAU/mL vs. 2081.0 [IQR 1077.0-3960.0] BAU/mL, respectively; p = 0.01). One participant who was initially seronegative in the control group remained seronegative after the booster dose. These findings suggest humoral immune response benefits of switching the standard immunosuppressive regimen to the regimen of mTORi, TAC, and prednisolone in KTRs during vaccination.
Collapse
Affiliation(s)
- Athiphat Banjongjit
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Supitchaya Phirom
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Jeerath Phannajit
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Division of Clinical Epidemiology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watsamon Jantarabenjakul
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Leilani Paitoonpong
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wonngarm Kittanamongkolchai
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Mahachakri Sirindhorn Clinical Research Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Salin Wattanatorn
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | | | - Somchai Eiam-Ong
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Yingyos Avihingsanon
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pokrath Hansasuta
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Jakapat Vanichanan
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Natavudh Townamchai
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Renal Immunology and Renal Transplant Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Excellence Center for Solid Organ Transplantation, King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
- Correspondence:
| |
Collapse
|
4
|
Saharia KK, Husson JS, Niederhaus SV, Iraguha T, Avila SV, Yoo YJ, Hardy NM, Fan X, Omili D, Crane A, Carrier A, Xie WY, Vander Mause E, Hankey K, Bauman S, Lesho P, Mannuel HD, Ahuja A, Mathew M, Avruch J, Baddley J, Goloubeva O, Shetty K, Dahiya S, Rapoport AP, Luetkens T, Atanackovic D. Humoral immunity against SARS-CoV-2 variants including omicron in solid organ transplant recipients after three doses of a COVID-19 mRNA vaccine. Clin Transl Immunology 2022; 11:e1391. [PMID: 35505864 PMCID: PMC9052011 DOI: 10.1002/cti2.1391] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives Solid organ transplant recipients (SOTR) receiving post‐transplant immunosuppression show increased COVID‐19‐related mortality. It is unclear whether an additional dose of COVID‐19 vaccines can overcome the reduced immune responsiveness against severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) variants. Methods We analysed humoral immune responses against SARS‐CoV‐2 and its variants in 53 SOTR receiving SARS‐CoV‐2 vaccination. Results Following the initial vaccination series, 60.3% of SOTR showed no measurable neutralisation and only 18.9% demonstrated neutralising activity of > 90%. More intensive immunosuppression, antimetabolites in particular, negatively impacted antiviral immunity. While absolute IgG levels were lower in SOTR than controls, antibody titres against microbial recall antigens were higher. By contrast, SOTR showed reduced vaccine‐induced IgG/IgA antibody titres against SARS‐CoV‐2 and its delta variants and fewer linear B‐cell epitopes, indicating reduced B‐cell diversity. Importantly, a third vaccine dose led to an increase in anti‐SARS‐CoV‐2 antibody titres and neutralising activity across alpha, beta and delta variants and to the induction of anti‐SARS‐CoV‐2 CD4+ T cells in a subgroup of patients analysed. By contrast, we observed significantly lower antibody titres after the third dose with the omicron variant compared to the ancestral SARS‐CoV‐2 and the improvement in neutralising activity was much less pronounced than for all the other variants. Conclusion Only a small subgroup of solid organ transplant recipients is able to generate functional antibodies after an initial vaccine series; however, an additional vaccine dose resulted in dramatically improved antibody responses against all SARS‐CoV‐2 variants except omicron where antibody responses and neutralising activity remained suboptimal.
Collapse
Affiliation(s)
- Kapil K Saharia
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA.,Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA
| | - Jennifer S Husson
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA.,Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA
| | - Silke V Niederhaus
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - Thierry Iraguha
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Stephanie V Avila
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Youngchae J Yoo
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA
| | - Nancy M Hardy
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Xiaoxuan Fan
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Destiny Omili
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Alice Crane
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - Amber Carrier
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - Wen Y Xie
- University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Department of Surgery University of Florida College of Medicine Gainesville FL USA
| | - Erica Vander Mause
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Kim Hankey
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Sherri Bauman
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Patricia Lesho
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Heather D Mannuel
- University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Baltimore Veterans Affairs Medical Center Baltimore MD USA
| | - Ashish Ahuja
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Minu Mathew
- Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA
| | - James Avruch
- Department of Surgery University of Maryland School of Medicine Baltimore MD USA
| | - John Baddley
- Institute of Human Virology University of Maryland School of Medicine Baltimore MD USA.,Divison of Infectious Diseases University of Maryland School of Medicine Baltimore MD USA.,University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Olga Goloubeva
- Department of Epidemiology and Public Health University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Kirti Shetty
- Division of Hepatology/Liver Transplantation University of Maryland School of Medicine Baltimore MD USA
| | - Saurabh Dahiya
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Aaron P Rapoport
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA
| | - Tim Luetkens
- Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Department of Microbiology and Immunology University of Maryland Baltimore MD USA
| | - Djordje Atanackovic
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Transplant and Cellular Therapy Program University of Maryland Greenebaum Comprehensive Cancer Center Baltimore MD USA.,Department of Microbiology and Immunology University of Maryland Baltimore MD USA
| |
Collapse
|
5
|
Magicova M, Zahradka I, Fialova M, Neskudla T, Gurka J, Modos I, Hojny M, Raska P, Smejkal P, Striz I, Viklicky O. Determinants of Immune Response to Anti-SARS-CoV-2 mRNA Vaccines in Kidney Transplant Recipients: A Prospective Cohort Study. Transplantation 2022; 106:842-852. [PMID: 34999659 PMCID: PMC8942601 DOI: 10.1097/tp.0000000000004044] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination has been recently shown to be impaired in kidney transplant recipients (KTRs), but the underlying factors affecting vaccine effectiveness need to be further elucidated. METHODS In this prospective cohort study, antibodies against S1 and S2 subunits of SARS-CoV-2 were evaluated using an immunochemiluminescent assay (cutoff 9.5 AU/mL, sensitivity 91.2%, and specificity 90.2%) in 736 KTRs, who were previously either naive or infected with SARS-CoV-2 and vaccinated before or after transplantation. Cellular response was analyzed in a subset of patients using an interferon gamma release assay (cutoff 0.15 IU/mL, sensitivity 92%, and specificity 100%). RESULTS Seroconversion was significantly more impaired in SARS-CoV-2-naive KTRs than in those previously infected (40.1% versus 97.1%; P < 0.001). Mycophenolate use (odds ratio, 0.15; 95% confidence interval, 0.09-0.24; P < 0.001) and depleting therapy in the past year (odds ratio, 0.19; 95% confidence interval, 0.05-0.8; P = 0.023) were found to be among independent factors associated with impaired humoral response. Similarly, the interferon gamma release assay tested in 50 KTRs (cutoff 0.15 IU/mL, sensitivity 92%, specificity 100%) showed that specific T-cell responses against spike protein epitopes are impaired in SARS-CoV-2-naive KTRs, as compared to previously infected KTRs (9.4% versus 90%, P < 0.001). All 35 KTRs vaccinated on the waiting list before transplantation exhibited sustained seroconversion persisting after transplantation. CONCLUSIONS Survivors of coronavirus disease 2019 and those vaccinated while on the waiting list exhibited a marked immune response to mRNA vaccines, contrary to poor response in naive KTRs vaccinated after transplantation (NCT04832841).
Collapse
Affiliation(s)
- Maria Magicova
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ivan Zahradka
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martina Fialova
- Department of Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Tomas Neskudla
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jiri Gurka
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Istvan Modos
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michal Hojny
- Institutional Pharmacy, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Raska
- Information Technology Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Smejkal
- Division of Hygiene and Epidemiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ilja Striz
- Department of Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ondrej Viklicky
- Department of Nephrology, Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Transplantation Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
6
|
Schuller M, Pfeifer V, Kirsch AH, Klötzer KA, Mooslechner AA, Rosenkranz AR, Stiegler P, Schemmer P, Sourij H, Eller P, Prietl B, Eller K. B Cell Composition Is Altered After Kidney Transplantation and Transitional B Cells Correlate With SARS-CoV-2 Vaccination Response. Front Med (Lausanne) 2022; 9:818882. [PMID: 35187002 PMCID: PMC8847739 DOI: 10.3389/fmed.2022.818882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The COVID-19 pandemic has major implications on kidney transplant recipients (KTRs) since they show increased mortality due to impaired immune responses to SARS-CoV-2 infection and a reduced efficacy of SARS-CoV-2 vaccination. Surprisingly, dialysis patients have shown superior seroconversion rates after vaccination compared to KTRs. Therefore, we investigated peripheral blood B cell (BC) composition before and after kidney transplantation (KT) and aimed to screen the BC compartment to explain impaired antibody generation. METHODS A total of 105 patients were recruited, and multicolor flow cytometric phenotyping of peripheral venous blood BC subpopulations was performed before and 1 year after KT. Complete follow-up was available for 71 individuals. Anti-SARS-CoV-2 antibodies were collected retrospectively and were available for 40 subjects, who had received two doses of an mRNA-based vaccine (BNT162b2 or mRNA-1273). RESULTS Overall, relative BC frequencies within lymphocytes decreased, and their absolute counts trended in the same direction 1 year after KT as compared to CKD G5 patients. Frequencies and absolute numbers of naïve BCs remained stable. Frequencies of double negative BCs, a heterogeneous subpopulation of antigen experienced BCs lacking CD27 expression, were increased after KT, yet their absolute counts were similar at both time points. Transitional BCs (TrBCs) and plasmablasts were significantly reduced after KT in absolute and relative terms. Memory BCs were affected differently since class-switched and IgM-only subsets decreased after KT, but unswitched and IgD-only memory BCs remained unchanged. CD86+ and CD5+ expression on BCs was downregulated after KT. Correlational analysis revealed that TrBCs were the only subset to correlate with titer levels after SARS-CoV-2 vaccination. Responders showed higher TrBCs, both absolute and relative, than non-responders. CONCLUSION Together, after 1 year, KTRs showed persistent and profound compositional changes within the BC compartment. Low TrBCs, 1 year after KT, may account for the low serological response to SARS-CoV-2 vaccination in KTRs compared to dialysis patients. Our findings need confirmation in further studies as they may guide vaccination strategies.
Collapse
Affiliation(s)
- Max Schuller
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Verena Pfeifer
- Center for Biomarker Research in Medicine, CBmed GmbH, Graz, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Alexander H. Kirsch
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Konstantin A. Klötzer
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Agnes A. Mooslechner
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Alexander R. Rosenkranz
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp Stiegler
- General, Visceral, and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Peter Schemmer
- General, Visceral, and Transplant Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp Eller
- Intensive Care Unit, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Barbara Prietl
- Center for Biomarker Research in Medicine, CBmed GmbH, Graz, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Kathrin Eller
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
7
|
D'Offizi G, Agrati C, Visco-Comandini U, Castilletti C, Puro V, Piccolo P, Montalbano M, Meschi S, Tartaglia E, Sorace C, Leone S, Lapa D, Grassi G, Goletti D, Ippolito G, Vaia F, Ettorre GM, Lionetti R. Coordinated cellular and humoral immune responses after two-dose SARS-CoV2 mRNA vaccination in liver transplant recipients. Liver Int 2022; 42:180-186. [PMID: 34719107 PMCID: PMC8662049 DOI: 10.1111/liv.15089] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 12/26/2022]
Abstract
Limited data are available on risks and benefits of anti-SARS-CoV2 vaccination in solid organ transplant recipients, and weaker responses have been described. At the Italian National Institute for Infectious Diseases, 61 liver transplant recipients underwent testing to describe the dynamics of humoral and cell-mediated immune response after two doses of anti-SARS-CoV2 mRNA vaccines and compared with 51 healthy controls. Humoral response was measured by quantifying both anti-spike and neutralizing antibodies; cell-mediated response was measured by PBMC proliferation assay with IFN-γ and IL-2 production. Liver transplant recipients showed lower response rates compared with controls in both humoral and cellular arms; shorter time since transplantation and multi-drug immunosuppressive regimen containing mycophenolate mofetil were predictive of reduced response to vaccination. Specific antibody and cytokine production, though reduced, were highly correlated in transplant recipients.
Collapse
Affiliation(s)
- Gianpiero D'Offizi
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Chiara Agrati
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | | | | | - Vincenzo Puro
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Paola Piccolo
- Internal Medicine, Fatebenefratelli Hospital Isola Tiberina, Rome
| | - Marzia Montalbano
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Silvia Meschi
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Eleonora Tartaglia
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Chiara Sorace
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Sara Leone
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Daniele Lapa
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Germana Grassi
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Delia Goletti
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | - Francesco Vaia
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| | | | - Raffaella Lionetti
- National Institute for Infectious Diseases L. Spallanzani - IRCCS (INMI) Rome
| |
Collapse
|
8
|
Duni A, Markopoulos GS, Mallioras I, Pappas H, Pappas E, Koutlas V, Tzalavra E, Baxevanos G, Priska S, Gartzonika K, Mitsis M, Dounousi E. The Humoral Immune Response to BNT162b2 Vaccine Is Associated With Circulating CD19+ B Lymphocytes and the Naïve CD45RA to Memory CD45RO CD4+ T Helper Cells Ratio in Hemodialysis Patients and Kidney Transplant Recipients. Front Immunol 2021; 12:760249. [PMID: 34925330 PMCID: PMC8678464 DOI: 10.3389/fimmu.2021.760249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022] Open
Abstract
Background The humoral and cellular immune responses to SARS-COV-2 vaccination remain to be elucidated in hemodialysis (HD) patients and kidney transplant recipients (KTRs), considering their baseline immunosuppressed status. The aim of our study was to assess the associations of vaccine-induced antibody responses with circulating lymphocytes sub-populations and their respective patterns of alterations in maintenance HD patients and KTRs. Materials and Methods We included 34 HD patients and 54 KTRs who received two doses of the mRNA-vaccine BNT162b2. Lymphocyte subpopulations were analyzed by flow cytometry before vaccination (T0), before the second vaccine dose (T1) and 2 weeks after the second dose (T2). The anti-SARS-CoV2 antibody response was assessed at T1 and at T2. Results 31 HD patients (91.8%) and 16 KTRs (29.6%) became seropositive at T2. HD patients who became seropositive following the first dose displayed higher CD19+ B lymphocytes compared to their seronegative HD counterparts. A positive correlation was established between CD19+ B cells counts and antibody titers at all time-points in both groups (p < 0.001). KTRs showed higher naïve CD4+CD45RA+ T helper cells compared to HD patients at baseline and T2 whereas HD patients displayed higher memory CD45RO+ T cells compared to KTRs at T2. The naïve CD4+CD45RA to memory CD4+CD45RO+ T helper cells fraction was negatively associated with antibody production in both groups. Conclusions Our study provides a potential conceptual framework for monitoring vaccination efficacy in HD patients and KTRs considering the correlation established between CD19+ B cells, generation of memory CD4+ T helper cells and anti SARS-CoV2 antibody response to vaccination.
Collapse
Affiliation(s)
- Anila Duni
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece.,Department of Surgery and Kidney Transplant Unit, University Hospital of Ioannina, Ioannina, Greece
| | - Georgios S Markopoulos
- Laboratory of Hematology - Unit of Molecular Biology, University Hospital of Ioannina, Ioannina, Greece
| | - Ioannis Mallioras
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | - Haralampos Pappas
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece.,Department of Surgery and Kidney Transplant Unit, University Hospital of Ioannina, Ioannina, Greece
| | | | - Vasileios Koutlas
- Department of Surgery and Kidney Transplant Unit, University Hospital of Ioannina, Ioannina, Greece
| | - Eirini Tzalavra
- Department of Surgery and Kidney Transplant Unit, University Hospital of Ioannina, Ioannina, Greece
| | - Gerasimos Baxevanos
- Laboratory of Hematology - Unit of Molecular Biology, University Hospital of Ioannina, Ioannina, Greece.,Internal Medicine Department, Hatzikosta General Hospital of Ioannina, Ioannina, Greece
| | - Silvia Priska
- Department of Nephrology, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Konstantina Gartzonika
- Microbiology Laboratory, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Michael Mitsis
- Department of Surgery and Kidney Transplant Unit, University Hospital of Ioannina, Ioannina, Greece
| | - Evangelia Dounousi
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece.,Department of Surgery and Kidney Transplant Unit, University Hospital of Ioannina, Ioannina, Greece.,Department of Nephrology, School of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
9
|
Curtis JR, Johnson SR, Anthony DD, Arasaratnam RJ, Baden LR, Bass AR, Calabrese C, Gravallese EM, Harpaz R, Kroger A, Sadun RE, Turner AS, Williams EA, Mikuls TR. American College of Rheumatology Guidance for COVID-19 Vaccination in Patients With Rheumatic and Musculoskeletal Diseases: Version 3. Arthritis Rheumatol 2021; 73:e60-e75. [PMID: 34346564 PMCID: PMC8426685 DOI: 10.1002/art.41928] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/10/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To provide guidance to rheumatology providers on the use of coronavirus disease 2019 (COVID-19) vaccines for patients with rheumatic and musculoskeletal diseases (RMDs). METHODS A task force was assembled that included 9 rheumatologists/immunologists, 2 infectious disease specialists, and 2 public health physicians. After agreeing on scoping questions, an evidence report was created that summarized the published literature and publicly available data regarding COVID-19 vaccine efficacy and safety, as well as literature for other vaccines in RMD patients. Task force members rated their agreement with draft consensus statements on a 9-point numerical scoring system, using a modified Delphi process and the RAND/University of California Los Angeles Appropriateness Method, with refinement and iteration over 2 sessions. Consensus was determined based on the distribution of ratings. RESULTS Despite a paucity of direct evidence, 74 draft guidance statements were developed by the task force and agreed upon with consensus to provide guidance for use of the COVID-19 vaccines in RMD patients and to offer recommendations regarding the use and timing of immunomodulatory therapies around the time of vaccination. CONCLUSION These guidance statements, made in the context of limited clinical data, are intended to provide direction to rheumatology health care providers on how to best use COVID-19 vaccines and to facilitate implementation of vaccination strategies for RMD patients.
Collapse
Affiliation(s)
| | - Sindhu R. Johnson
- Toronto Western HospitalMount Sinai Hospital, and University of TorontoTorontoOntarioCanada
| | - Donald D. Anthony
- Louis Stokes Cleveland VA Medical CenterMetroHealth Medical Center, and Case Western Reserve UniversityClevelandOhioUnited States
| | - Reuben J. Arasaratnam
- VA North Texas Health Care System and University of Texas Southwestern Medical CenterDallas
| | | | - Anne R. Bass
- Hospital for Special Surgery and Weill Cornell MedicineNew YorkNew YorkUnited States
| | | | | | - Rafael Harpaz
- Harpaz Herman ConsultantsAtlantaGeorgiaUnited States
| | | | | | - Amy S. Turner
- American College of RheumatologyAtlantaGeorgiaUnited States
| | | | - Ted R. Mikuls
- University of Nebraska Medical Center and VA Nebraska–Western Iowa Health Care SystemOmaha
| |
Collapse
|
10
|
Diks AM, Overduin LA, van Leenen LD, Slobbe L, Jolink H, Visser LG, van Dongen JJM, Berkowska MA. B-Cell Immunophenotyping to Predict Vaccination Outcome in the Immunocompromised - A Systematic Review. Front Immunol 2021; 12:690328. [PMID: 34557188 PMCID: PMC8452967 DOI: 10.3389/fimmu.2021.690328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
Vaccination is the most effective measure to prevent infections in the general population. Its efficiency strongly depends on the function and composition of the immune system. If the immune system lacks critical components, patients will not be fully protected despite a completed vaccination schedule. Antigen-specific serum immunoglobulin levels are broadly used correlates of protection. These are the products of terminally differentiated B cells - plasma cells. Here we reviewed the literature on how aberrancies in B-cell composition and function influence immune responses to vaccinations. In a search through five major literature databases, 6,537 unique articles published from 2000 and onwards were identified. 75 articles were included along three major research lines: extremities of life, immunodeficiency and immunosuppression. Details of the protocol can be found in the International Prospective Register of Systematic Reviews [PROSPERO (registration number CRD42021226683)]. The majority of articles investigated immune responses in adults, in which vaccinations against pneumococci and influenza were strongly represented. Lack of baseline information was the most common reason of exclusion. Irrespective of study group, three parameters measured at baseline seemed to have a predictive value in assessing vaccine efficacy: (1) distribution of B-cell subsets (mostly a reduction in memory B cells), (2) presence of exhausted/activated B cells, or B cells with an aberrant phenotype, and (3) pre-existing immunological memory. In this review we showed how pre-immunization (baseline) knowledge of circulating B cells can be used to predict vaccination efficacy. We hope that this overview will contribute to optimizing vaccination strategies, especially in immunocompromised patients.
Collapse
Affiliation(s)
- Annieck M Diks
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Lisanne A Overduin
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Laurens D van Leenen
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Lennert Slobbe
- Department of Internal Medicine, Section of Infectious Diseases, Institute for Tropical Diseases, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Hetty Jolink
- Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Leonardus G Visser
- Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - Magdalena A Berkowska
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| |
Collapse
|
11
|
Curtis JR, Johnson SR, Anthony DD, Arasaratnam RJ, Baden LR, Bass AR, Calabrese C, Gravallese EM, Harpaz R, Kroger A, Sadun RE, Turner AS, Williams EA, Mikuls TR. American College of Rheumatology Guidance for COVID-19 Vaccination in Patients With Rheumatic and Musculoskeletal Diseases: Version 2. Arthritis Rheumatol 2021; 73:e30-e45. [PMID: 34128356 PMCID: PMC8427105 DOI: 10.1002/art.41877] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To provide guidance to rheumatology providers on the use of coronavirus disease 2019 (COVID-19) vaccines for patients with rheumatic and musculoskeletal diseases (RMDs). METHODS A task force was assembled that included 9 rheumatologists/immunologists, 2 infectious disease specialists, and 2 public health physicians. After agreeing on scoping questions, an evidence report was created that summarized the published literature and publicly available data regarding COVID-19 vaccine efficacy and safety, as well as literature for other vaccines in RMD patients. Task force members rated their agreement with draft consensus statements on a 9-point numerical scoring system, using a modified Delphi process and the RAND/University of California Los Angeles Appropriateness Method, with refinement and iteration over 2 sessions. Consensus was determined based on the distribution of ratings. RESULTS Despite a paucity of direct evidence, 74 draft guidance statements were developed by the task force and agreed upon with consensus to provide guidance for use of the COVID-19 vaccines in RMD patients and to offer recommendations regarding the use and timing of immunomodulatory therapies around the time of vaccination. CONCLUSION These guidance statements, made in the context of limited clinical data, are intended to provide direction to rheumatology health care providers on how to best use COVID-19 vaccines and to facilitate implementation of vaccination strategies for RMD patients.
Collapse
Affiliation(s)
| | - Sindhu R. Johnson
- Toronto Western HospitalMount Sinai Hospital, and University of TorontoTorontoOntarioCanada
| | - Donald D. Anthony
- Louis Stokes Cleveland VA Medical CenterMetroHealth Medical Center, and Case Western Reserve UniversityClevelandOhioUnited States
| | - Reuben J. Arasaratnam
- VA North Texas Health Care System and University of Texas Southwestern Medical CenterDallas
| | | | - Anne R. Bass
- Hospital for Special Surgery and Weill Cornell MedicineNew YorkNew YorkUnited States
| | | | | | - Rafael Harpaz
- Harpaz Herman ConsultantsAtlantaGeorgiaUnited States
| | | | | | - Amy S. Turner
- American College of RheumatologyAtlantaGeorgiaUnited States
| | | | - Ted R. Mikuls
- University of Nebraska Medical Center and VA Nebraska–Western Iowa Health Care SystemOmaha
| |
Collapse
|
12
|
Gresham LM, Marzario B, Dutz J, Kirchhof MG. An evidence-based guide to SARS-CoV-2 vaccination of patients on immunotherapies in dermatology. J Am Acad Dermatol 2021; 84:1652-1666. [PMID: 33482251 PMCID: PMC7816618 DOI: 10.1016/j.jaad.2021.01.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022]
Abstract
Immune-mediated diseases and immunotherapeutics can negatively affect normal immune functioning and, consequently, vaccine safety and response. The COVID-19 pandemic has incited research aimed at developing a novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine. As SARS-CoV-2 vaccines are developed and made available, the assessment of anticipated safety and efficacy in patients with immune-mediated dermatologic diseases and requiring immunosuppressive and/or immunomodulatory therapy is particularly important. A review of the literature was conducted by a multidisciplinary committee to provide guidance on the safety and efficacy of SARS-CoV-2 vaccination for dermatologists and other clinicians when prescribing immunotherapeutics. The vaccine platforms being used to develop SARS-CoV-2 vaccines are expected to be safe and potentially effective for dermatology patients on immunotherapeutics. Current guidelines for the vaccination of an immunocompromised host remain appropriate when considering future administration of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Louise M Gresham
- Division of Dermatology, Department of Medicine, University of Ottawa and The Ottawa Hospital, Ottawa, Canada
| | - Barbara Marzario
- Division of Dermatology, Department of Medicine, University of Ottawa and The Ottawa Hospital, Ottawa, Canada
| | - Jan Dutz
- Department of Dermatology and Skin Sciences, University of British Columbia, Vancouver, Canada
| | - Mark G Kirchhof
- Division of Dermatology, Department of Medicine, University of Ottawa and The Ottawa Hospital, Ottawa, Canada.
| |
Collapse
|
13
|
L'huillier AG, Ferreira VH, Hirzel C, Natori Y, Slomovic J, Ku T, Hoschler K, Ierullo M, Selzner N, Schiff J, Singer LG, Humar A, Kumar D. Cell-Mediated Immune Responses After Influenza Vaccination of Solid Organ Transplant Recipients: Secondary Outcomes Analyses of a Randomized Controlled Trial. J Infect Dis 2020; 221:53-62. [PMID: 31550354 DOI: 10.1093/infdis/jiz471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/12/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Despite annual immunization, solid organ transplant (SOT) patients remain at increased risk for severe influenza infection because of suboptimal vaccine immunogenicity. We aimed to compare the CD4+ and CD8+ T-cell responses of the high-dose (HD) and the standard-dose (SD) trivalent inactivated vaccine. METHODS We collected peripheral blood mononuclear cells pre- and postimmunization from 60 patients enrolled in a randomized trial of HD versus SD vaccine (30 HD; 30 SD) during the 2016-2017 influenza season. RESULTS The HD vaccine elicited significantly greater monofunctional and polyfunctional CD4+ and CD8+ T-cell responses against influenza A/H1N1, A/H3N2, and B. For example, median vaccine-elicited influenza-specific polyfunctional CD4+ T cells were higher in recipients of the HD than SD vaccine after stimulation with influenza A/H1N1 (1193 vs 0 per 106 CD4+ T cells; P = .003), A/H3N2 (1154 vs 51; P = .008), and B (1102 vs 0; P = .001). Likewise, vaccine-elicited influenza-specific polyfunctional CD8+ T cells were higher in recipients of the HD than SD vaccine after stimulation with influenza B (367 vs 0; P = .002). CONCLUSIONS Our study provides novel evidence that HD vaccine elicits greater cellular responses compared with the SD vaccine in SOT recipients, which provides support to preferentially consider use of HD vaccination in the SOT setting.
Collapse
Affiliation(s)
| | - Victor H Ferreira
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Cedric Hirzel
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Yoichiro Natori
- Division of Infectious Disease, Department of Medicine, University of Miami Miller School of Medicine and Miami Transplant Institute, Miami, Florida, USA
| | - Jaclyn Slomovic
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Terrance Ku
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | | | - Matthew Ierullo
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Nazia Selzner
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Jeffrey Schiff
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Lianne G Singer
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Atul Humar
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| | - Deepali Kumar
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada
| |
Collapse
|
14
|
Natori Y, Shiotsuka M, Slomovic J, Hoschler K, Ferreira V, Ashton P, Rotstein C, Lilly L, Schiff J, Singer L, Humar A, Kumar D. A Double-Blind, Randomized Trial of High-Dose vs Standard-Dose Influenza Vaccine in Adult Solid-Organ Transplant Recipients. Clin Infect Dis 2019; 66:1698-1704. [PMID: 29253089 DOI: 10.1093/cid/cix1082] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/13/2017] [Indexed: 12/21/2022] Open
Abstract
Background The annual standard-dose (SD) influenza vaccine has suboptimal immunogenicity in solid organ transplant recipients (SOTRs). Influenza vaccine that contains higher doses of antigens may lead to greater immunogenicity in this population. Methods We conducted a randomized, double-blind trial to compare the safety and immunogenicity of the 2016-2017 high-dose (HD; FluzoneHD, Sanofi) vs SD (Fluviral, GSK) influenza vaccine in adult SOTRs. Preimmunization and 4-week postimmunization sera underwent strain-specific hemagglutination inhibition assay. Results We enrolled 172 patients who received study vaccine, and 161 (84 HD; 77 SD) were eligible for analysis. Seroconversion to at least 1 of 3 vaccine antigens was present in 78.6% vs 55.8% in HD vs SD vaccine groups (P < .001), respectively. Seroconversions to A/ H1N1, A/H3N2, and B strains were 40.5% vs 20.5%, 57.1% vs 32.5%, and 58.3% vs 41.6% in HD vs SD vaccine groups (P = .006, P = .002, P = .028, respectively). Post-immunization geometric mean titers of A/H1N1, A/H3N2, and B strains were significantly higher in the HD group (P = .007, P = .002, P = .033). Independent factors associated with seroconversion to at least 1 vaccine strain were the use of HD vaccine (odds ratio [OR], 3.23; 95% confidence interval [CI], 1.56-6.67) and use of mycophenolate doses <2 g daily (OR, 2.76; 95% CI, 1.12-6.76). Conclusions HD vaccine demonstrated significantly better immunogenicity than SD vaccine in adult transplant recipients and may be the preferred influenza vaccine for this population. Clinical Trials Registration NCT03139565.
Collapse
Affiliation(s)
- Yoichiro Natori
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Mika Shiotsuka
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Jaclyn Slomovic
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | | | - Victor Ferreira
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Peter Ashton
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Coleman Rotstein
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Les Lilly
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Jeffrey Schiff
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Lianne Singer
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Atul Humar
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| | - Deepali Kumar
- Multi Organ Transplant Program, University Health Network, University of Toronto, Ontario, Canada
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW The aim of this study was to highlight recent evidence on important aspects of influenza vaccination in solid organ transplant recipients. RECENT FINDINGS Influenza vaccine is the most evaluated vaccine in transplant recipients. The immunogenicity of the vaccine is suboptimal after transplantation. Newer formulations such as inactivated unadjuvanted high-dose influenza vaccine and the administration of a booster dose within the same season have shown to increase response rates. Intradermal vaccination and adjuvanted vaccines did not show clear benefit over standard influenza vaccines. Recent studies in transplant recipients do not suggest a higher risk for allograft rejection, neither after vaccination with a standard influenza vaccine nor after the administration of nonstandard formulation (high-dose, adjuvanted vaccines), routes (intradermally) or a booster dose. Nevertheless, influenza vaccine coverage in transplant recipients is still unsatisfactory low, potentially due to misinterpretation of risks and benefits. SUMMARY Annual influenza vaccination is well tolerated and is an important part of long-term care of solid organ transplant recipients.
Collapse
|
16
|
Luukkainen A, Puan KJ, Yusof N, Lee B, Tan KS, Liu J, Yan Y, Toppila-Salmi S, Renkonen R, Chow VT, Rotzschke O, Wang DY. A Co-culture Model of PBMC and Stem Cell Derived Human Nasal Epithelium Reveals Rapid Activation of NK and Innate T Cells Upon Influenza A Virus Infection of the Nasal Epithelium. Front Immunol 2018; 9:2514. [PMID: 30467502 PMCID: PMC6237251 DOI: 10.3389/fimmu.2018.02514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/11/2018] [Indexed: 12/30/2022] Open
Abstract
Background: We established an in vitro co-culture model involving H3N2-infection of human nasal epithelium with peripheral blood mononuclear cells (PBMC) to investigate their cross-talk during early H3N2 infection. Methods: Nasal epithelium was differentiated from human nasal epithelial stem/progenitor cells and cultured wtih fresh human PBMC. PBMC and supernatants were harvested after 24 and 48 h of co-culture with H3N2-infected nasal epithelium. We used flow cytometry and Luminex to characterize PBMC subpopulations, their activation and secretion of cytokine and chemokines. Results: H3N2 infection of the nasal epithelium associated with significant increase in interferons (IFN-α, IFN-γ, IL-29), pro-inflammatory cytokines (TNF-α, BDNF, IL-3) and viral-associated chemokines (IP-10, MCP-3, I-TAC, MIG), detectable already after 24 h. This translates into rapid activation of monocytes, NK-cells and innate T-cells (MAIT and γδ T cells), evident with CD38+ and/or CD69+ upregulation. Conclusions: This system may contribute to in vitro mechanistic immunological studies bridging systemic models and possibly enable the development of targeted immunomodulatory therapies.
Collapse
Affiliation(s)
- Annika Luukkainen
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Kia Joo Puan
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Nurhashikin Yusof
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Liu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yan Yan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sanna Toppila-Salmi
- Haartman Institute, University of Helsinki, Helsinki, Finland.,Skin and Allergy Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Risto Renkonen
- Haartman Institute, University of Helsinki, Helsinki, Finland.,HUSLAB, Helsinki University Hospital, Helsinki, Finland
| | - Vincent T Chow
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Olaf Rotzschke
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Esposito S, Bonanni P, Maggi S, Tan L, Ansaldi F, Lopalco PL, Dagan R, Michel JP, van Damme P, Gaillat J, Prymula R, Vesikari T, Mussini C, Frank U, Osterhaus A, Celentano LP, Rossi M, Guercio V, Gavazzi G. Recommended immunization schedules for adults: Clinical practice guidelines by the Escmid Vaccine Study Group (EVASG), European Geriatric Medicine Society (EUGMS) and the World Association for Infectious Diseases and Immunological Disorders (WAidid). Hum Vaccin Immunother 2016; 12:1777-94. [PMID: 27135390 PMCID: PMC4964839 DOI: 10.1080/21645515.2016.1150396] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 01/19/2016] [Accepted: 02/01/2016] [Indexed: 12/27/2022] Open
Abstract
Rapid population aging has become a major challenge in the industrialized world and progressive aging is a key reason for making improvement in vaccination a cornerstone of public health strategy. An increase in age-related disorders and conditions is likely to be seen in the near future, and these are risk factors for the occurrence of a number of vaccine-preventable diseases. An improvement in infectious diseases prevention specifically aimed at adults and the elderly can therefore also decrease the burden of these chronic conditions by reducing morbidity, disability, hospital admissions, health costs, mortality rates and, perhaps most importantly, by improving the quality of life. Among adults, it is necessary to identify groups at increased risk of vaccine-preventable diseases and highlight the epidemiological impact and benefits of vaccinations using an evidence-based approach. This document provides clinical practice guidance on immunization for adults in order to provide recommendations for decision makers and healthcare workers in Europe. Although immunization is considered one of the most impactful and cost-effective public health measures that can be undertaken, vaccination coverage rates among adults are largely lower than the stated goal of ≥ 95% among adults, and stronger efforts are needed to increase coverage in this population. Active surveillance of adult vaccine-preventable diseases, determining the effectiveness of the vaccines approved for marketing in the last 5 y, the efficacy and safety of vaccines in immunocompromised patients, as well as in pregnant women, represent the priorities for future research.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Highly Intensive Care Unit, University of Milan, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Bonanni
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Stefania Maggi
- CNR - Institute of Neuroscience, Aging Branch Center for Research, Padua, Italy
| | - Litjan Tan
- Immunization Action Coalition, St Paul, MN, USA
| | - Filippo Ansaldi
- Department of Health Sciences, University of Genoa, IRCCS San Martino-IST University Teaching Hospital, Genoa, Italy
| | | | - Ron Dagan
- Pediatric Infectious Disease Unit, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University, Beer-Sheva, Israel
| | | | - Pierre van Damme
- Vaccine & Infectious Disease Institute, Antwerp University, Wilrijk, Belgium
| | | | - Roman Prymula
- Department of Social Medicine, Faculty of Medicine, Charles University, Sokolska, Hradec Kralove, Czech Republic
| | - Timo Vesikari
- Vaccine Research Center, Tampere University Hospital, Tampere, Finland
| | - Cristina Mussini
- Clinic of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Uwe Frank
- Division of Infection Control and Hospital Epidemiology, Department of Infectious Diseases, Heidelberg University, Heidelberg, Germany
| | | | | | - Marta Rossi
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Valentina Guercio
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Gaetan Gavazzi
- University Clinic of Geriatric Medicine, University Hospital of Grenoble, and GREPI University of Grenoble-Alpes, Grenoble, France
| |
Collapse
|
18
|
Héquet D, Pascual M, Lartey S, Pathirana RD, Bredholt G, Hoschler K, Hullin R, Meylan P, Cox RJ, Manuel O. Humoral, T-cell and B-cell immune responses to seasonal influenza vaccine in solid organ transplant recipients receiving anti-T cell therapies. Vaccine 2016; 34:3576-83. [PMID: 27219339 DOI: 10.1016/j.vaccine.2016.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/29/2016] [Accepted: 05/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND We analyzed the impact of the anti-T-cell agents basiliximab and antithymocyte globulins (ATG) on antibody and cell-mediated immune responses after influenza vaccination in solid-organ transplant recipients. METHODS 71 kidney and heart transplant recipients (basiliximab [n=43] and ATG [n=28]) received the trivalent influenza vaccine. Antibody responses were measured at baseline and 6 weeks post-vaccination by hemagglutination inhibition assay; T-cell responses were measured by IFN-γ ELISpot assays and intracellular cytokine staining (ICS); and influenza-specific memory B-cell (MBC) responses were evaluated using ELISpot. RESULTS Median time of vaccination from transplantation was 29 months (IQR 8-73). Post-vaccination seroconversion rates were 26.8% for H1N1, 34.1% for H3N2 and 4.9% for influenza B in the basiliximab group and 35.7% for H1N1, 42.9% for H3N2 and 14.3% for influenza B in the ATG group (p=0.44, p=0.61, and p=0.21, respectively). The number of influenza-specific IFN-γ-producing cells increased significantly after vaccination (from 35 to 67.5 SFC/10(6) PBMC, p=0.0007), but no differences between treatment groups were observed (p=0.88). Median number of IgG-MBC did not increase after vaccination (H1N1, p=0.94; H3N2 p=0.34; B, p=0.79), irrespective of the type of anti-T-cell therapy. CONCLUSIONS After influenza vaccination, a significant increase in antibody and T-cell immune responses but not in MBC responses was observed in transplant recipients. Immune responses were not significantly different between groups that received basiliximab or ATG.
Collapse
Affiliation(s)
- Delphine Héquet
- Transplantation Center, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Infectious Diseases Service, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Manuel Pascual
- Transplantation Center, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Sarah Lartey
- Influenza Centre, Department of Clinical Science, University of Bergen, Norway
| | - Rishi D Pathirana
- Influenza Centre, Department of Clinical Science, University of Bergen, Norway
| | - Geir Bredholt
- Influenza Centre, Department of Clinical Science, University of Bergen, Norway
| | - Katja Hoschler
- Public Health England, Microbiology Services Colindale, London, United Kingdom
| | - Roger Hullin
- Division of Cardiology, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Pascal Meylan
- Infectious Diseases Service, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Institute of Microbiology, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Rebecca J Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Norway; Department of Research and Development, Haukeland University Hospital, Bergen, Norway; Jebsen Centre for Influenza Vaccine Research, University of Bergen, Norway
| | - Oriol Manuel
- Transplantation Center, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Infectious Diseases Service, University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Demir Z, Frange P, Lacaille F. Vaccinations, response, and controls before and after intestinal transplantation in children. Pediatr Transplant 2016; 20:449-55. [PMID: 26847771 DOI: 10.1111/petr.12669] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2015] [Indexed: 11/25/2022]
Abstract
Vaccination is an effective strategy to decrease infections in transplant recipients. Children after intestinal transplantation carry a high risk of infection due to increased immunosuppression. In a series of 22 children after intestinal transplantation, we studied the vaccination schedules and the antibodies against vaccine-preventable diseases before transplantation, and at one and five yr after transplantation. We reviewed whether the vaccination schedules were complete, and we analysed the factors that may influence serological immunity and the incidence of disease in patients with deficient immunity. All patients completed the recommended vaccination schedules for DTaP-IPV and HBV. After transplantation, the negative antibodies against vaccine-preventable diseases were mostly related to an antirejection therapy: for DTaP-IPV: four of four patients with no antibody had been treated for rejection, for HBV: two of five, HAV: three of four, MMR: three of seven, and VZV: three of four. A post-transplantation varicella infection was followed by acute rejection, with probability for a relationship between both events. We observed 50% of varicella cases in unvaccinated children, highlighting the importance of pretransplant vaccination. Waning immunogenicity mediated by antibodies against vaccine-preventable disease after transplantation indicated a need for boosters. The recommendations should be regularly enforced, as the reliance on routine immunizations schedules is not adequate in immunocompromised patients.
Collapse
Affiliation(s)
- Z Demir
- Pediatric Hepato-Gastro-Enterology-Nutrition Unit, Necker-Enfants Malades Hospital, Paris, France
| | - P Frange
- Microbiology Department, Necker-Enfants Malades Hospital, Paris, France.,Pediatric Immunology and Hematology Unit, Necker Hospital, Paris, France.,EA 7327, Paris Descartes University, Sorbonne Paris Cite, Paris, France
| | - F Lacaille
- Pediatric Hepato-Gastro-Enterology-Nutrition Unit, Necker-Enfants Malades Hospital, Paris, France
| |
Collapse
|
20
|
Spies C, Luetz A, Lachmann G, Renius M, von Haefen C, Wernecke KD, Bahra M, Schiemann A, Paupers M, Meisel C. Influence of Granulocyte-Macrophage Colony-Stimulating Factor or Influenza Vaccination on HLA-DR, Infection and Delirium Days in Immunosuppressed Surgical Patients: Double Blind, Randomised Controlled Trial. PLoS One 2015; 10:e0144003. [PMID: 26641243 PMCID: PMC4671639 DOI: 10.1371/journal.pone.0144003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 11/11/2015] [Indexed: 11/19/2022] Open
Abstract
PURPOSE Surgical patients are at high risk for developing infectious complications and postoperative delirium. Prolonged infections and delirium result in worse outcome. Granulocyte-macrophage colony-stimulating factor (GM-CSF) and influenza vaccination are known to increase HLA-DR on monocytes and improve immune reactivity. This study aimed to investigate whether GM-CSF or vaccination reverses monocyte deactivation. Secondary aims were whether it decreases infection and delirium days after esophageal or pancreatic resection over time. METHODS In this prospective, randomized, placebo-controlled, double-blind, double dummy trial setting on an interdisciplinary ICU of a university hospital 61 patients with immunosuppression (monocytic HLA-DR [mHLA-DR] <10,000 monoclonal antibodies [mAb] per cell) on the first day after esophageal or pancreatic resection were treated with either GM-CSF (250 μg/m2/d), influenza vaccination (Mutagrip 0.5 ml/d) or placebo for a maximum of 3 consecutive days if mHLA-DR remained below 10,000 mAb per cell. HLA-DR on monocytes was measured daily until day 5 after surgery. Infections and delirium were followed up for 9 days after surgery. Primary outcome was HLA-DR on monocytes, and secondary outcomes were duration of infection and delirium. RESULTS mHLA-DR was significantly increased compared to placebo (p < 0.001) and influenza vaccination (p < 0.001) on the second postoperative day. Compared with placebo, GM-CSF-treated patients revealed shorter duration of infection (p < 0.001); the duration of delirium was increased after vaccination (p = 0.003). CONCLUSION Treatment with GM-CSF in patients with postoperative immune suppression was safe and effective in restoring monocytic immune competence. Furthermore, therapy with GM-CSF reduced duration of infection in immune compromised patients. However, influenza vaccination increased duration of delirium after major surgery. TRIAL REGISTRATION www.controlled-trials.com ISRCTN27114642.
Collapse
Affiliation(s)
- Claudia Spies
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
- * E-mail:
| | - Alawi Luetz
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Gunnar Lachmann
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Markus Renius
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | | | - Marcus Bahra
- Department of General, Abdominal and Transplantation Surgery, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Alexander Schiemann
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Marco Paupers
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Christian Meisel
- Institute of Medical Immunology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| |
Collapse
|