20 results on '"Böhmig, Georg A."'
Search Results
2. Natural Killer Cell Presence in Antibody-Mediated Rejection.
- Author
-
Diebold, Matthias, Farkash, Evan A., Barnes, Jenna, Regele, Heinz, Kozakowski, Nicolas, Schatzl, Martina, Mayer, Katharina A., Haindl, Susanne, Vietzen, Hannes, Hidalgo, Luis G., Halloran, Philip F., Eskandary, Farsad, and Böhmig, Georg A.
- Subjects
KILLER cells ,GRAFT rejection ,KIDNEY transplantation ,GRAFT survival ,GENETIC variation - Abstract
Transcript analyses highlight an important contribution of natural killer (NK) cells to microvascular inflammation (MVI) in antibody-mediated rejection (ABMR), but only few immunohistologic studies have quantified their spatial distribution within graft tissue. This study included 86 kidney transplant recipients who underwent allograft biopsies for a positive donor-specific antibody (DSA) result. NK cells were visualized and quantified within glomeruli and peritubular capillaries (PTC), using immunohistochemistry for CD34 alongside CD16/T-bet double-staining. Staining results were analyzed in relation to histomorphology, microarray analysis utilizing the Molecular Microscope Diagnostic System, functional NK cell genetics, and clinical outcomes. The number of NK cells in glomeruli per mm2 glomerular area (NKglom) and PTC per mm2 cortical area (NKPTC) was substantially higher in biopsies with ABMR compared to those without rejection, and correlated with MVI scores (NKglom Spearman's correlation coefficient [SCC] = 0.55, p < 0.001, NKPTC 0.69, p < 0.001). In parallel, NK cell counts correlated with molecular classifiers reflecting ABMR activity (ABMRprob: NKglom 0.59, NKPTC 0.75) and showed a trend towards higher levels in association with high functional FCGR3A and KLRC2 gene variants. Only NKPTC showed a marginally significant association with allograft function and survival. Our immunohistochemical results support the abundance of NK cells in DSApositive ABMR. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Determinants of the intercept and slope of glomerular filtration rate in recipients of a live donor kidney transplant
- Author
-
Hamböck, Martina, Staudenherz, Anton, Kainz, Alexander, Geist, Barbara, Hecking, Manfred, Doberer, Konstantin, Hacker, Marcus, and Böhmig, Georg A.
- Published
- 2021
- Full Text
- View/download PDF
4. Morphologic and Molecular Features of Antibody-Mediated Transplant Rejection: Pivotal Role of Molecular Injury as an Independent Predictor of Renal Allograft Functional Decline.
- Author
-
Herz, Carsten T., Diebold, Matthias, Kainz, Alexander, Mayer, Katharina A., Doberer, Konstantin, Kozakowski, Nicolas, Halloran, Philip F., and Böhmig, Georg A.
- Subjects
GRAFT rejection ,KIDNEY failure ,HOMOGRAFTS ,KIDNEY diseases ,GLOMERULAR filtration rate ,WOUNDS & injuries ,GRAFT survival - Abstract
Current knowledge about the factors correlating with functional decline and subsequent failure of kidney allografts in antibody-mediated rejection (ABMR) is limited. We conducted a cohort study involving 75 renal allograft recipients diagnosed with late ABMR occurring at least 6 months after transplantation. The study aimed to examine the correlation of molecular and histologic features with estimated glomerular filtration rate (eGFR) trajectories and death-censored graft survival. We focused on sum scores reflecting histologic ABMR activity versus chronicity and molecular scores of ABMR probability (ABMR
Prob ), injury-repair response (IRRAT) and fibrosis (ciprob). In multivariable Cox analysis, a Banff lesion-based chronicity index (ci+ct+cg[x2]; hazard ratio per interquartile range [IQR]: 1.97 [95% confidence interval: 0.97 to 3.99]) and IRRAT (1.93 [0.96 to 3.89]) showed the strongest associations with graft failure. Among biopsy variables, IRRAT exhibited the highest relative variable importance and emerged as the sole independent predictor of eGFR slope (change per IQR: −4.2 [−7.8 to −0.6] mL/ min/1.73 m² /year). In contrast, morphologic chronicity associated with baseline eGFR only. We conclude that the extent of molecular injury is a robust predictor of renal function decline. Transcriptome analysis has the potential to improve outcome prediction and possibly identify modifiable injury, guiding targeted therapeutic interventions. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
5. Morphologic and Molecular Features of Antibody-Mediated Transplant Rejection: Pivotal Role of Molecular Injury as an Independent Predictor of Renal Allograft Functional Decline.
- Author
-
Herz, Carsten T., Diebold, Matthias, Kainz, Alexander, Mayer, Katharina A., Doberer, Konstantin, Kozakowski, Nicolas, Halloran, Philip F., and Böhmig, Georg A.
- Subjects
GRAFT rejection ,KIDNEY failure ,HOMOGRAFTS ,KIDNEY diseases ,GLOMERULAR filtration rate ,WOUNDS & injuries ,GRAFT survival - Abstract
Current knowledge about the factors correlating with functional decline and subsequent failure of kidney allografts in antibody-mediated rejection (ABMR) is limited. We conducted a cohort study involving 75 renal allograft recipients diagnosed with late ABMR occurring at least 6 months after transplantation. The study aimed to examine the correlation of molecular and histologic features with estimated glomerular filtration rate (eGFR) trajectories and death-censored graft survival. We focused on sum scores reflecting histologic ABMR activity versus chronicity and molecular scores of ABMR probability (ABMRProb), injury-repair response (IRRAT) and fibrosis (ciprob). In multivariable Cox analysis, a Banff lesion-based chronicity index (ci+ct+cg[x2]; hazard ratio per interquartile range [IQR]: 1.97 [95% confidence interval: 0.97 to 3.99]) and IRRAT (1.93 [0.96 to 3.89]) showed the strongest associations with graft failure. Among biopsy variables, IRRAT exhibited the highest relative variable importance and emerged as the sole independent predictor of eGFR slope (change per IQR: -4.2 [-7.8 to -0.6] mL/min/1.73m2/year). In contrast, morphologic chronicity associated with baseline eGFR only. We conclude that the extent of molecular injury is a robust predictor of renal function decline. Transcriptome analysis has the potential to improve outcome prediction and possibly identify modifiable injury, guiding targeted therapeutic interventions. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Post-Transplant Surveillance and Management of Chronic Active Antibody-Mediated Rejection in Renal Transplant Patients in Europe.
- Author
-
Rostaing, Lionel P. E., Böhmig, Georg A., Gibbons, Ben, and Taqi, Muhammed Mahdi
- Subjects
- *
KIDNEY transplantation , *GRAFT rejection , *DELAYED diagnosis , *DIAGNOSIS , *PHYSICIANS , *BK virus - Abstract
Antibody mediated rejection (ABMR) is the leading cause of immune-related allograft failure following kidney transplantation. Chronic active ABMR (CABMR) typically occurs after one-year post-transplant andis themost common causeof late allograft failure. This studywas designed to assess common practices in Europe for post-transplant surveillance 1year after kidney transplant, as well as the diagnosis and management of CABMR. A 15-minute online survey with 58 multiple choice or open-ended questions was completed by EU transplant nephrologists, transplant surgeons and nephrologists. Survey topics included patient caseloads, post-transplant routine screening and treatment of CABMR. The results indicated that observing clinical measures of graft function form the cornerstone of posttransplant surveillance. This may be suboptimal, leading to late diagnoses and untreatable disease. Indeed, less than half of patients who develop CABMR receive treatment beyond optimization of immune suppression. This is attributable to not only late diagnoses, but also a lack of proven efficacious therapies. Intravenous Immunoglobulin (IVIG), steroid pulse and apheresis are prescribed by the majority to treat CABMR. While biologics can feature as part of treatment, there is no single agent that is being used by more than half of physicians. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Diagnosis and treatment of antibody-mediated kidney allograft rejection
- Author
-
Böhmig, Georg and Regele, Heinz
- Published
- 2003
- Full Text
- View/download PDF
8. Deletion of the Natural Killer Cell Receptor NKG2C Encoding KLR2C Gene and Kidney Transplant Outcome.
- Author
-
Vietzen, Hannes, Döhler, Bernd, Tran, Thuong Hien, Süsal, Caner, Halloran, Philip F., Eskandary, Farsad, Herz, Carsten T., Mayer, Katharina A., Kozakowski, Nicolas, Wahrmann, Markus, Ely, Sarah, Haindl, Susanne, Puchhammer-Stöckl, Elisabeth, and Böhmig, Georg A.
- Subjects
KILLER cell receptors ,KILLER cells ,KIDNEY transplantation ,GRAFT rejection ,TREATMENT effectiveness - Abstract
Natural killer (NK) cells may contribute to antibody-mediated rejection (ABMR) of renal allografts. The role of distinct NK cell subsets in this specific context, such as NK cells expressing the activating receptor NKG2C, is unknown. Our aim was to investigate whether KLRC2 gene deletion variants which determine NKG2C expression affect the pathogenicity of donor-specific antibodies (DSA) and, if so, influence long-term graft survival. We genotyped the KLRC2
wt/del variants for two distinct kidney transplant cohorts, (i) a cross-sectional cohort of 86 recipients who, on the basis of a positive post-transplant DSA result, all underwent allograft biopsies, and (ii) 1,860 recipients of a deceased donor renal allograft randomly selected from the Collaborative Transplant Study (CTS) database. In the DSA+ patient cohort, KLRC2wt/wt (80%) was associated with antibody-mediated rejection (ABMR; 65% versus 29% among KLRC2wt/del subjects; P =0.012), microvascular inflammation [MVI; median g+ptc score: 2 (interquartile range: 0-4) versus 0 (0-1), P =0.002], a molecular classifier of ABMR [0.41 (0.14-0.72) versus 0.10 (0.07-0.27), P =0.001], and elevated NK cell-related transcripts (P =0.017). In combined analyses of KLRC2 variants and a functional polymorphism in the Fc gamma receptor IIIA gene (FCGR3A -V/F158), ABMR rates and activity gradually increased with the number of risk genotypes. In DSA+ and CTS cohorts, however, the KLRC2wt/wt variant did not impact long-term death-censored graft survival, also when combined with the FCGR3A- V158 risk variant. KLRC2wt/wt may be associated with DSA-triggered MVI and ABMR-associated gene expression patterns, but the findings observed in a highly selected cohort of DSA+ patients did not translate into meaningful graft survival differences in a large multicenter kidney transplant cohort not selected for HLA sensitization. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
9. Diagnostic value of donor‐derived cell‐free DNA to predict antibody‐mediated rejection in donor‐specific antibody‐positive renal allograft recipients.
- Author
-
Mayer, Katharina A., Doberer, Konstantin, Tillgren, Amanda, Viard, Thierry, Haindl, Susanne, Krivanec, Sebastian, Reindl‐Schwaighofer, Roman, Eder, Michael, Eskandary, Farsad, Casas, Silvia, Wahrmann, Markus, Regele, Heinz, and Böhmig, Georg A.
- Subjects
CELL-free DNA ,RECEIVER operating characteristic curves ,ALLOIMMUNITY - Abstract
Summary: Circulating donor‐specific antibodies (DSA) do not necessarily indicate antibody‐mediated rejection (ABMR). Here, we evaluated the diagnostic value of donor‐derived cell‐free DNA (dd‐cfDNA) as an add‐on to DSA detection. The study included two independent cohorts of DSA+ kidney allograft recipients, 45 subclinical cases identified by cross‐sectional antibody screening (cohort 1), and 30 recipients subjected to indication biopsies (cohort 2). About 50% of the DSA+ recipients had ABMR and displayed higher dd‐cfDNA levels than DSA+ABMR− recipients (cohort 1: 1.90% [median; IQR: 0.78–3.90%] vs. 0.52% [0.35–0.72%]; P < 0.001); (cohort 2: 1.20% [0.82–2.50%] vs. 0.59% [0.28–2.05%]; P = 0.086). Receiver operating characteristic (ROC) analysis revealed an area under the curve (AUC) of 0.89 and 0.69 for dd‐cfDNA, and 0.88 and 0.77 for DSA mean fluorescence intensity (MFI), respectively. In combined models, adding dd‐cfDNA to DSA‐MFI or vice versa significantly improved the diagnostic accuracy. Limited diagnostic performance of dd‐cfDNA in cohort 2 was related to the frequent finding of other types of graft injury among ABMR− recipients, like T cell‐mediated rejection or glomerulonephritis. For dd‐cfDNA in relation to injury of any cause an AUC of 0.97 was calculated. Monitoring of dd‐cfDNA in DSA+ patients may be a useful tool to detect ABMR and other types of injury. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Functional Fc Gamma Receptor Gene Polymorphisms and Long-Term Kidney Allograft Survival.
- Author
-
Wahrmann, Markus, Döhler, Bernd, Arnold, Marie-Luise, Scherer, Sabine, Mayer, Katharina A., Haindl, Susanne, Haslacher, Helmuth, Böhmig, Georg A., and Süsal, Caner
- Subjects
GRAFT survival ,FC receptors ,GENETIC polymorphisms ,GRAFT rejection ,TREATMENT effectiveness - Abstract
The functional Fc gamma receptor (FcγR) IIIA polymorphism FCGR3A -V/F158 was earlier suggested to determine the potential of donor-specific HLA antibodies to trigger microcirculation inflammation, a key lesion of antibody-mediated renal allograft rejection. Associations with long-term transplant outcomes, however, have not been evaluated to date. To clarify the impact of FCGR3A -V/F158 polymorphism on kidney transplant survival, we genotyped a cohort of 1,940 recipient/donor pairs. Analyzing 10-year death-censored allograft survival, we found no significant differences in relation to FCGR3A -V/F158. There was also no independent survival effect in a multivariable Cox model. Similarly, functional polymorphisms in two other activating FcγR, FCGR2A -H/R131 (FcγRIIA) and FCGR3B -NA1/NA2 (FcγRIIIB), were not associated with outcome. There were also no significant survival differences among patient subgroups at increased risk of rejection-related injury, such as pre-sensitized recipients (> 0% panel reactivity; n = 438) or recipients treated for rejection within the first year after transplantation (n = 229). Our study results suggest that the earlier shown association of FcγR polymorphism with microcirculation inflammation may not be strong enough to exert a meaningful effect on graft survival. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
11. Anti‐interleukin‐6 antibody clazakizumab in late antibody‐mediated kidney transplant rejection: effect on cytochrome P450 drug metabolism.
- Author
-
Mühlbacher, Jakob, Schörgenhofer, Christian, Doberer, Konstantin, Dürr, Michael, Budde, Klemens, Eskandary, Farsad, Mayer, Katharina A., Schranz, Sabine, Ely, Sarah, Reiter, Birgit, Chong, Edward, Adler, Scott H., Jilma, Bernd, and Böhmig, Georg A.
- Subjects
DRUG metabolism ,CYTOCHROME P-450 ,GRAFT rejection ,KIDNEY transplantation ,INFLAMMATION - Abstract
Summary: Targeting interleukin‐6 (IL‐6) is a promising strategy to counteract antibody‐mediated rejection (ABMR). In inflammatory states, IL‐6 antagonism was shown to modulate cytochrome P450 (CYP), but its impact on drug metabolism in ABMR treatment was not addressed so far. We report a sub‐study of a phase 2 trial of anti‐IL‐6 antibody clazakizumab in late ABMR (ClinicalTrials.gov, NCT03444103). Twenty kidney transplant recipients were randomized to clazakizumab versus placebo (4‐weekly doses; 12 weeks), followed by a 9‐month extension where all recipients received clazakizumab. To study CYP2C19/CYP3A4 metabolism, we administered pantoprazole (20 mg intravenously) at prespecified time points. Dose‐adjusted C0 levels (C0/D ratio) of tacrolimus (n = 13) and cyclosporin A (CyA, n = 6) were monitored at 4‐weekly intervals. IL‐6 and C‐reactive protein were not elevated at baseline, the latter was then suppressed to undetectable levels under clazakizumab. IL‐6 blockade had no clinically meaningful impact on pantoprazole pharmacokinetics (area under the curve; baseline versus week 52: 3.16 [2.21–7.84] versus 4.22 [1.99–8.18] μg/ml*h, P = 0.36) or calcineurin inhibitor C0/D ratios (tacrolimus: 1.49 [1.17–3.20] versus 1.37 [0.98–2.42] ng/ml/mg, P = 0.21; CyA: 0.69 [0.57–0.85] versus 1.08 [0.52–1.38] ng/ml/mg, P = 0.47). We conclude that IL‐6 blockade in ABMR – in absence of systemic inflammation – may have no meaningful effect on CYP metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
12. Renal allograft DARCness in subclinical acute and chronic active ABMR.
- Author
-
Kläger, Johannes, Eskandary, Farsad, Böhmig, Georg A., Kozakowski, Nicolas, Kainz, Alexander, Colin Aronovicz, Yves, Cartron, Jean‐Pierre, Segerer, Stephan, and Regele, Heinz
- Subjects
GENE expression profiling ,ANTIGEN receptors ,GRAFT survival ,DIAGNOSTIC immunohistochemistry ,DIAGNOSIS - Abstract
Summary: Gene expression profiling of renal allograft biopsies revealed the Duffy antigen receptor for chemokines (DARC) as being strikingly upregulated in antibody‐mediated rejection (ABMR). DARC has previously been shown to be associated with endothelial injury. This study aimed at assessing the value of DARC immunohistochemistry as diagnostic marker in ABMR. The study was performed on 82 prospectively collected biopsies of a clinically well‐defined population (BORTEJECT trial, NCT01873157) of DSA‐positive patients with gene expression data available for all biopsies. Diagnostic histologic assessment of biopsies was performed according to the Banff diagnostic scheme. DARC expression was focally accentuated, on peritubular capillaries (PTC) mostly in areas of interstitial fibrosis and/or inflammation. DARC positivity was associated with diagnosis of ABMR and correlated with DARC gene expression levels detected by microarray analysis. Still, as previously described, a substantial number of biopsies without signs of rejection showed DARC‐positive PTC. We did not observe significantly reduced graft survival in cases showing histologic signs of ABMR and being DARC‐positive, as compared to DARC‐negative ABMR. In summary, the upregulation of DARC, detected by immunohistochemistry, is associated with but not specific for ABMR. We did not observe reduced graft survival in DARC‐positive patients. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
13. Outcome of Extracorporeal Photopheresis as an Add-On Therapy for Antibody-Mediated Rejection in Lung Transplant Recipients.
- Author
-
Benazzo, Alberto, Worel, Nina, Schwarz, Stefan, Just, Ulrike, Nechay, Anna, Lambers, Christoph, Böhmig, Georg, Fischer, Gottfried, Koren, Daniela, Muraközy, Gabriela, Knobler, Robert, Klepetko, Walter, Hoetzenecker, Konrad, and Jaksch, Peter
- Abstract
Introduction: The diagnosis and treatment of antibody-mediated rejection (AMR) after lung transplantation has recently gained recognition within the transplant community. Extracorporeal photopheresis (ECP), currently used to treat chronic lung allograft dysfunction, modulates various pathways of the immune system known to be involved in AMR. We hypothesize that adding ECP to established AMR treatments could prevent the rebound of donor-specific antibodies (DSA). Objectives: This study aimed to analyze the role of ECP as an add-on therapy to prevent the rebound of DSA. Methods: Lung transplant recipients who received ECP as an add-on therapy for pulmonary AMR between January 2010 and January 2019 were included in this single-center retrospective analysis. Baseline demographics of the patients, as well as their immunological characteristics and long-term transplant outcomes, were analyzed. Results: A total of 41 patients developed clinical AMR during the study period. Sixteen patients received ECP as an add-on therapy after first-line AMR treatment. Among the 16 patients, 2 (13%) had pretransplant DSA, both against human leukocyte antigen (HLA) class I (B38, B13, and C06). Fifteen patients (94%) developed de novo DSA (dnDSA), i.e., 10 (63%) against class I and 14 (88%) against class II. The median time to dnDSA after lung transplantation was 361 days (range 25–2,548). According to the most recent International Society of Heart and Lung Transplantation (ISHLT) consensus report, 2 (13%) patients had definite clinical AMR, 6 (38%) had probable AMR, and 7 (44%) had possible AMR. The median mean fluorescence intensity (MFI) of dnDSA at the time of clinical diagnosis was 4,220 (range 1,319–10,552) for anti-HLA class I and 10,953 (range 1,969–27,501) for anti-HLA class II antibodies. ECP was performed for a median of 14 cycles (range 1–64). MFI values of dnDSA against HLA classes I and II were significantly reduced over the treatment period (for anti-class I: 752; range 70–2,066; for anti-class II: 5,612; range 1,689–21,858). The 1-year survival rate was 55%. No adverse events related to ECP were reported in any of the patients. Conclusions: ECP is associated with a reduction of dnDSA in lung transplant recipients affected by AMR. Prospective studies are warranted to confirm the beneficial effects of ECP in the setting of AMR. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
14. The therapeutic challenge of late antibody‐mediated kidney allograft rejection.
- Author
-
Böhmig, Georg A., Eskandary, Farsad, Doberer, Konstantin, and Halloran, Philip F.
- Subjects
- *
IMMUNOGLOBULIN producing cells , *PLASMA cells , *PROTEASOME inhibitors , *B cells , *KIDNEY failure - Abstract
Summary: Late antibody‐mediated rejection (ABMR) is a cardinal cause of kidney allograft failure, manifesting as a continuous and, in contrast with early rejection, often clinically silent alloimmune process. While significant progress has been made towards an improved understanding of its molecular mechanisms and the definition of diagnostic criteria, there is still no approved effective treatment. In recent small randomized controlled trials, therapeutic strategies with promising results in observational studies, such as proteasome inhibitor bortezomib, anti‐C5 antibody eculizumab, or high dose intravenous immunoglobulin plus rituximab, had no significant impact in late and/or chronic ABMR. Such disappointing results reinforce a need of new innovative treatment strategies. Potential candidates may be the interference with interleukin‐6 to modulate B cell alloimmunity, or innovative compounds that specifically target antibody‐producing plasma cells, such as antibodies against CD38. Given the phenotypic heterogeneity of ABMR, the design of adequate systematic trials to assess the safety and efficiency of such therapies, however, is challenging. Several trials are currently being conducted, and new developments will hopefully provide us with effective ways to counteract the deleterious impact of antibody‐mediated graft injury. Meanwhile, the weight of evidence would suggest that, when approaching using existing treatments for established antibody‐mediated rejection, "less may be more". [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
15. Deceased donor kidney transplantation across donor-specific antibody barriers: predictors of antibody-mediated rejection.
- Author
-
Schwaiger, Elisabeth, Eskandary, Farsad, Kozakowski, Nicolas, Bond, Gregor, Kikić, Željko, Yoo, Daniel, Rasoul-Rockenschaub, Susanne, Oberbauer, Rainer, and Böhmig, Georg A.
- Subjects
KIDNEY transplantation ,IMMUNOGLOBULINS ,GRAFT rejection ,IMMUNOLOGY ,HEMAPHERESIS - Abstract
Background. Apheresis-based desensitization allows for successful transplantation across major immunological barriers. For donor-specific antibody (DSA)- and/or crossmatchpositive transplantation, however, it has been shown that even intense immunomodulation may not completely prevent antibody- mediated rejection (ABMR). Methods. In this study, we evaluated transplant outcomes in 101 DSA+ deceased donor kidney transplant recipients (transplantation between 2009 and 2013; median follow-up: 24 months) who were subjected to immunoadsorption (IA)- based desensitization. Treatment included a single pretransplant IA session, followed by anti-lymphocyte antibody and serial post-transplant IA. In 27 cases, a positive complement- dependent cytotoxicity crossmatch (CDCXM) was rendered negative immediately before transplantation. Seventy-four of the DSA+ recipients had a negative CDCXM already before IA. Results. Three-year death-censored graft survival in DSA+ patients was significantly worse than in 513 DSA- recipients transplanted during the same period (79 versus 88%, P = 0.008). Thirty-three DSA+ recipients (33%) had ABMR.While a positive baseline CDCXM showed only a trend towards higher ABMR rates (41 versus 30% in CDCXM- recipients, P = 0.2), DSA mean fluorescence intensity (MFI) in single bead assays significantly associated with rejection, showing 20 versus 71% ABMR rates at <5000 versus >15 000 peak DSA MFI. The predictive value of MFI was moderate, with the highest accuracy at a median of 13 300 MFI (after cross-validation: 0.72). Other baseline variables, including CDC assay results, human leukocyte antigen mismatch, prior transplantation or type of induction treatment, did not add independent predictive information. Conclusions. IA-based desensitization failed to prevent ABMR in a considerable number of DSA+ recipients. Assessing DSA MFI may help stratify risk of rejection, supporting its use as a guide to organ allocation and individualized treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
16. Complement inhibition as potential new therapy for antibody-mediated rejection.
- Author
-
Eskandary, Farsad, Wahrmann, Markus, Mühlbacher, Jakob, and Böhmig, Georg A.
- Subjects
GRAFT rejection ,KIDNEY transplantation ,HOMOGRAFTS ,IMMUNOGLOBULINS ,ECULIZUMAB - Abstract
Antibody-mediated rejection ( ABMR) is a leading cause of kidney allograft failure. While the exact mechanisms contributing to donor-specific antibody ( DSA)-triggered tissue injury are still incompletely understood, complement activation via the classical pathway is believed to be one of the key players. There is now growing interest in complement blockade as an antirejection treatment. One attractive strategy may be inhibition of terminal complex formation using anti-C5 antibody eculizumab. Anecdotal reports, case series, and a unique cohort of flow crossmatch-positive live donor kidney transplant recipients subjected to eculizumab-based desensitization have demonstrated successful prevention and reversal of acute clinical ABMR. Nevertheless, maybe due to complement activation steps proximal of C5 or even complement-independent mechanisms, subclinical rejection processes that might culminate in chronic injury were found to escape inhibition. Larger studies designed to clarify the actual clinical value of terminal complement inhibition as an antirejection treatment are currently underway. In addition, alternative concepts, such as therapies that target key component C1, are currently under development, and we will see in the near future whether new strategies in the pipeline will have the potential to beneficially impact clinical practice. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
17. Detection of alloantibody-mediated complement activation: A diagnostic advance in monitoring kidney transplant rejection?
- Author
-
Böhmig, Georg A., Kikic, Zeljko, Wahrmann, Markus, Eskandary, Farsad, Aliabadi, Arezu Z., Zlabinger, Gerhard J., Regele, Heinz, and Feucht, Helmut E.
- Subjects
- *
IMMUNOGLOBULINS , *KIDNEY transplantation , *GRAFT rejection , *PHENOTYPES , *CELL-mediated cytotoxicity , *HLA histocompatibility antigens - Abstract
Objective Antibody-mediated rejection (ABMR) is an important cause of kidney allograft injury. In the last two decades, detection of complement split product C4d along transplant capillaries, a footprint of antibody-mediated classical complement activation, has evolved as a useful diagnostic marker of ABMR. While it was recognized that ABMR may occur also in the absence of C4d, numerous studies have shown that C4d deposition may indicate a more severe rejection phenotype associated with poor graft survival. Such studies suggest a possible diagnostic benefit of ex vivo monitoring the complement-activating capability of circulating alloantibodies. Design and methods We reviewed the literature between 1993 and 2015, focusing on in vivo (biopsy work-up) and in vitro detection (modified bead array technology) of HLA antibody-triggered classical complement activation in kidney transplantation. Results Precise HLA antibody detection methods, in particular Luminex-based single antigen bead (SAB) assays, have provided a valuable basis for the design of techniques for in vitro detection of HLA antibody-triggered complement activation reflected by C1q, C4 or C3 split product deposition to the bead surface. Establishing such assays it was recognized that deposition of complement products to SAB, which critically depends on antibody binding strength, may be a cardinal trigger of the prozone effect, a troublesome in vitro artifact caused by a steric interference with IgG detection reagents. False-low IgG results, especially on SAB with extensive antibody binding, have to be considered when interpreting studies analyzing the diagnostic value of complement in relation to standard IgG detection. Levels of complement-fixing donor-specific antibodies (DSA) were shown to correlate with the results of standard crossmatch tests, suggesting potential application for crossmatch prediction. Moreover, while the utility of pre-transplant complement detection, at least in crossmatch-negative transplant recipients, is controversially discussed, a series of studies have shown that the appearance of post-transplant complement-fixing DSA may be associated with C4d deposition in transplant capillaries and a particular risk of graft failure. Conclusions The independent value of modified single antigen bead assays, as compared to a careful analysis of standard IgG detection, which may be affected considerably by complement dependent artifacts, needs to be clarified. Whether they have the potential to improve the predictive accuracy of our current diagnostic repertoire warrants further study. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
18. Bortezomib in late antibody-mediated kidney transplant rejection (BORTEJECT Study): study protocol for a randomized controlled trial.
- Author
-
Eskandary, Farsad, Bond, Gregor, Schwaiger, Elisabeth, Kikic, Zeljko, Winzer, Christine, Wahrmann, Markus, Marinova, Lena, Haslacher, Helmuth, Regele, Heinz, Oberbauer, Rainer, and Böhmig, Georg A.
- Subjects
ORGAN donors ,BORTEZOMIB ,KIDNEY transplantation ,TRANSPLANTATION of organs, tissues, etc. ,OUTPATIENT medical care - Abstract
Background Despite major advances in transplant medicine, improvements in long-term kidney allograft survival have not been commensurate with those observed shortly after transplantation. The formation of donor-specific antibodies (DSA) and ongoing antibody-mediated rejection (AMR) processes may critically contribute to late graft loss. However, appropriate treatment for late AMR has not yet been defined. There is accumulating evidence that the proteasome inhibitor bortezomib may substantially affect the function and integrity of alloantibodysecreting plasma cells. The impact of this agent on the course of late AMR has not so far been systematically investigated. Methods/design The BORTEJECT Study is a randomized controlled trial designed to clarify the impact of intravenous bortezomib on the course of late AMR. In this single-center study (nephrological outpatient service, Medical University Vienna) we plan an initial cross-sectional DSA screening of 1,000 kidney transplant recipients (functioning graft at ≥180 days; estimated glomerular filtration rate (eGFR) >20 ml/minute/1.73 m²). DSA-positive recipients will be subjected to kidney allograft biopsy to detect morphological features consistent with AMR. Forty-four patients with biopsy-proven AMR will then be included in a double-blind placebocontrolled intervention trial (1:1 randomization stratified for eGFR and the presence of Tcell- mediated rejection). Patients in the active group will receive two cycles of bortezomib (4 × 1.3 mg/m² over 2 weeks; 3-month interval between cycles). The primary end point will be the course of eGFR over 24 months (intention-to-treat analysis). The sample size was calculated according to the assumption of a 5 ml/minute/1.73 m² difference in eGFR slope (per year) between the two groups (alpha: 0.05; power: 0.8). Secondary endpoints will be DSA levels, protein excretion, measured glomerular filtration rate, transplant and patient survival, and the development of acute and chronic morphological lesions in 24-month protocol biopsies. Discussion The impact of anti-humoral treatment on the course of late AMR has not yet been systematically investigated. Based on the hypothesis that proteasome inhibition improves the outcome of DSA-positive late AMR, we suggest that our trial has the potential to provide solid evidence towards the treatment of this type of rejection. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. Posttransplant Human Leukocyte Antigen Antibodies in Stable Kidney Transplant Recipients.
- Author
-
Bartel, Gregor and Böhmig, Georg A.
- Subjects
- *
KIDNEY transplantation , *GRAFT rejection , *HLA histocompatibility antigens - Abstract
Detection of donor-specific antibodies represents a major criterion of antibody-mediated rejection. This type of rejection, a leading cause of long-term allograft failure, may be the consequence of a multistep process initiated by the formation of donor-specific antibodies, which may subsequently trigger microcirculation inflammation and tissue damage followed by graft dysfunction and loss. At first sight this concept is in strong support of implementing a per-protocol longitudinal antibody monitoring in all kidney transplant recipients. One may speculate that early detection of donor-specific antibody occurrence could guide timely anti-humoral treatment, preventing subsequent irreversible graft damage. However, recent studies have revealed controversial results. In contrast to recipients with graft dysfunction, a considerable proportion of patients with normal function at the time of antibody testing were shown to maintain excellent long-term survival despite detectable de novo donor-specific antibodies. Moreover, the persistence of detectable antibodies following desensitization in immunological high-risk patients was described to be not necessarily associated with inferior transplant performance. For donor-specific antibody positive stable patients, a role of transplant accommodation, a state of acquired resistance to immune injury, was speculated. The present review focuses on the still controversial issue of donor-specific antibody monitoring in kidney transplant recipients, putting a special focus on stable patients who present without clinical signs of ongoing rejection. [ABSTRACT FROM AUTHOR]
- Published
- 2014
20. Clazakizumab in late antibody-mediated rejection: study protocol of a randomized controlled pilot trial.
- Author
-
Eskandary, Farsad, Dürr, Michael, Budde, Klemens, Doberer, Konstantin, Reindl-Schwaighofer, Roman, Waiser, Johannes, Wahrmann, Markus, Regele, Heinz, Spittler, Andreas, Lachmann, Nils, Firbas, Christa, Mühlbacher, Jakob, Bond, Gregor, Halloran, Philipp F., Chong, Edward, Jilma, Bernd, and Böhmig, Georg A.
- Subjects
IMMUNOGLOBULINS ,RANDOMIZED controlled trials ,RITUXIMAB ,INTERLEUKIN-6 ,IMMUNOSUPPRESSION - Abstract
Background: Late antibody-mediated rejection (ABMR) triggered by donor-specific antibodies (DSA) is a cardinal cause of kidney allograft dysfunction and loss. Diagnostic criteria for this rejection type are well established, but effective treatment remains a major challenge. Recent randomized controlled trials (RCT) have failed to demonstrate the efficacy of widely used therapies, such as rituximab plus intravenous immunoglobulin or proteasome inhibition (bortezomib), reinforcing a great need for new therapeutic concepts. One promising target in this context may be interleukin-6 (IL-6), a pleiotropic cytokine known to play an important role in inflammation and adaptive immunity.Methods: This investigator-driven RCT was designed to assess the safety and efficacy of clazakizumab, a genetically engineered humanized monoclonal antibody directed against IL-6. The study will include 20 DSA-positive kidney allograft recipients diagnosed with ABMR ≥ 365 days after transplantation. Participants will be recruited at two study sites in Austria and Germany (Medical University of Vienna; Charité University Medicine Berlin). First, patients will enter a three-month double-blind RCT (1,1 randomization, stratification according to ABMR phenotype and study site) and will receive either clazakizumab (subcutaneous administration of 25 mg in monthly intervals) or placebo. In a second open-label part of the trial (months 4-12), all patients will receive clazakizumab at 25 mg every month. The primary endpoint is safety and tolerability. Secondary endpoints are the pharmacokinetics and pharmacodynamics of clazakizumab, its effect on drug metabolism in the liver, DSA characteristics, morphological ABMR lesions and molecular gene expression patterns in three- and 12-month protocol biopsies, serum/urinary biomarkers of inflammation and endothelial activation/injury, Torque Teno viral load as a measure of overall immunosuppression, kidney function, urinary protein excretion, as well as transplant and patient survival.Discussion: Currently, there is no treatment proven to be effective in halting the progression of late ABMR. Based on the hypothesis that antagonizing the effects of IL-6 improves the outcome of DSA-positive late ABMR by counteracting DSA-triggered inflammation and B cell/plasma cell-driven alloimmunity, we suggest that our trial has the potential to provide proof of concept of a novel treatment of this type of rejection.Trial Registration: ClinicalTrials.gov, NCT03444103 . Registered on 23 February 2018 (retrospective registration). [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.