151
|
Urzykowska A, Piątosa B, Grycuk U, Kowalewski G, Kułaga Z, Grenda R. Evaluation of Cumulative Effect of Standard Triple Immunosuppression on Prevention of De Novo Donor Specific Antibodies (dnDSA) Production in Children after Kidney Transplantation—A Retrospective and Prospective Study. CHILDREN 2021; 8:children8121162. [PMID: 34943360 PMCID: PMC8700537 DOI: 10.3390/children8121162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022]
Abstract
De novo Donor Specific Antibodies (dnDSA) are associated with inferior graft outcomes. Standard immunosuppression is expected to prevent dnDSA production in low-risk patients. We have evaluated a cumulative effect of a triple immunosuppression (CNI/MMF/Pred), as well as TAC concentration and coefficient of variation on the incidence of dnDSA production. Overall, 67 transplanted patients were evaluated in retrospective (dnDSA for-cause; n = 29) and prospective (dnDSA by protocol; n = 38) groups. In the retrospective group, the eGFR value at first dnDSA detection (median interval—4.0 years post-transplant) was 41 mL/min/1.73 m2; 55% of patients presented biopsy-proven cAMR, and 41% lost the graft within next 2.4 years. Patients from the prospective group presented 97% graft survival and eGFR of 76 mL/min/1.73 m2 at 2 years follow-up, an overall incidence of 21% of dnDSA and 18% of acute (T cell) rejection. None of the patients from the prospective group developed cAMR. Median value of Vasudev score within 2 years of follow-up was not significantly higher in dsDSA negative patients, while median value of TAC C0 > 1–24 months post-transplant was 7.9 in dnDSA negative vs. 7.1 ng/mL in dnDSA positive patients (p = 0.008). Conclusion: dnDSA-negative patients presented a higher exposure to tacrolimus, while not to the combined immunosuppression.
Collapse
Affiliation(s)
- Agnieszka Urzykowska
- Department of Nephrology, Kidney Transplantation & Hypertension, Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Barbara Piątosa
- Histocompatibility Laboratory, Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (B.P.); (U.G.)
| | - Urszula Grycuk
- Histocompatibility Laboratory, Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (B.P.); (U.G.)
| | - Grzegorz Kowalewski
- Department of Surgery and Organ Transplantation, Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Zbigniew Kułaga
- Department of Public Health, Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
| | - Ryszard Grenda
- Department of Nephrology, Kidney Transplantation & Hypertension, Children’s Memorial Health Institute, 04-730 Warsaw, Poland;
- Correspondence:
| |
Collapse
|
152
|
Novotny M, Hruba P, Kment M, Voska L, Kabrtova K, Slavcev A, Viklicky O. Intimal Arteritis and Microvascular Inflammation Are Associated With Inferior Kidney Graft Outcome, Regardless of Donor-Specific Antibodies. Front Med (Lausanne) 2021; 8:781206. [PMID: 34957155 PMCID: PMC8692297 DOI: 10.3389/fmed.2021.781206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/03/2021] [Indexed: 11/21/2022] Open
Abstract
Background: The prognostic role of intimal arteritis of kidney allografts in donor-specific antibody negative (DSA–) antibody-mediated rejection (ABMR) remains unclear. Methods: Seventy-two out of 881 patients who had undergone kidney transplantation from 2014 to 2017 exhibited intimal arteritis in biopsies performed during the first 12 months. In 26 DSA negative cases, the intimal arteritis was accompanied by tubulointerstitial inflammation as part of T cell-mediated vascular rejection (TCMRV, N = 26); intimal arteritis along with microvascular inflammation occurred in 29 DSA negative (ABMRV/DSA–) and 19 DSA positive cases (ABMRV, DSA+, N = 17). In 60 (83%) patients with intimal arteritis, the surveillance biopsies after antirejection therapy were performed. Hundred and two patients with non-vascular ABMR with DSA (ABMR/DSA+, N = 55) and without DSA (ABMR/DSA–, N = 47) served as controls. Time to transplant glomerulopathy (TG) and graft failure were the study endpoints. Results: Transplant glomerulopathy -free survival at 36 months was 100% in TCMRV, 85% in ABMR/DSA–, 65% in ABMRV/DSA-, 54% in ABMR/DSA+ and 31% in ABMRV/DSA+ (log rank p < 0.001). Death-censored graft survival at 36 months was 98% in ABMR/DSA-, 96% in TCMRV, 86% in ABMRV/DSA–, 79% in ABMR/DSA+, and 64% in ABMRV/DSA+ group (log rank p = 0.001). In surveillance biopsies, the resolution of rejection was found in 19 (90%) TCMRV, 14 (58%) ABMRV/DSA–, and only 4 (27%) ABMRV/DSA+ patients (p = 0.006). In the multivariable model, intimal arteritis as part of ABMR represented a significant risk for TG development (HR 2.1, 95% CI 1.2–3.8; p = 0.012) regardless of DSA status but not for graft failure at 36 months. Conclusions: Intimal arteritis as part of ABMR represented a risk for early development of TG regardless of the presence or absence of DSA. Intimal arteritis in DSA positive ABMR represented the high-risk phenotype.
Collapse
Affiliation(s)
- Marek Novotny
- Department of Nephrology, Transplant Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
- Institute of Physiology, First Medical Faculty, Charles University, Prague, Czechia
| | - Petra Hruba
- Transplant Laboratory, Transplant Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Martin Kment
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Ludek Voska
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Katerina Kabrtova
- Department of Immunogenetics, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Antonij Slavcev
- Department of Immunogenetics, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Ondrej Viklicky
- Department of Nephrology, Transplant Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
- Institute of Physiology, First Medical Faculty, Charles University, Prague, Czechia
- Transplant Laboratory, Transplant Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
- *Correspondence: Ondrej Viklicky
| |
Collapse
|
153
|
Oblak M, Mlinšek G, Kojc N, Frelih M, Buturović-Ponikvar J, Arnol M. Spot Urine Protein Excretion in the First Year Following Kidney Transplantation Associates With Allograft Rejection Phenotype at 1-Year Surveillance Biopsies: An Observational National-Cohort Study. Front Med (Lausanne) 2021; 8:781195. [PMID: 34869503 PMCID: PMC8635090 DOI: 10.3389/fmed.2021.781195] [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: 09/22/2021] [Accepted: 10/13/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Urine protein excretion is routinely measured to assess kidney allograft injury, but the diagnostic value of this measurement for kidney transplant pathology remains unclear. Here we investigated whether spot urine protein excretion in the first year following transplantation associates with allograft rejection phenotype at 1-year surveillance biopsies and de-novo occurrence of donor-specific antibodies (DSA). Patients and Methods: This prospective, observational national-cohort study included 139 non-sensitized patients who received a deceased donor kidney transplant between December 2014 and 2018. All patients received basiliximab induction and tacrolimus-based immunosuppression. Estimated protein excretion rate (ePER) was calculated monthly from spot urine protein-to-creatinine ratios. At 1-year, all recipients underwent surveillance graft biopsy and were screened for de-novo DSA. Screening-positive sera were subjected to single antigen bead (SAB) testing. The occurrence of de-novo DSA was determined based on SAB reactivity patterns using a mean fluorescence intensity threshold >1,000. Results: Among the 139 study patients, 27 patients (19%) had histologic evidence of T cell-mediated rejection (TCMR), and 9 patients (7%) had histologic evidence of antibody-mediated rejection (AMR) at 1-year surveillance biopsy. One year after transplant, 19 patients (14%) developed de-novo DSA. Compared with patients without rejection and no de-novo DSA, mixed-effects linear regression analysis showed a significant difference in slope of ePER during the first year in patients with AMR and de-novo DSA at 1-year (46, 95% CI 25-68 mg/day/1.73 m2 per month and 34, 95% CI 20-49 mg/day/1.73 m2 per month, respectively). Patients with vascular TCMR also showed a significant difference in ePER slope over time compared with patients with non-rejection findings (31, 95% CI 9-52 mg/day/1.73 m2 per month). The discriminatory power of ePER for intragraft rejection processes was better in patients with AMR (AUC 0.95, 95% CI 0.90-0.99; P < 0.001) than in those with TCMR (AUC 0.68, 95% CI 0.59-0.79; P = 0.002), with 89% sensitivity and 93% specificity for proteinuria >550 mg/day/1.73m2. Conclusions: An increase in ePER in the first year following kidney transplantation associates with AMR, vascular TCMR and de-novo DSA at 1-year and may be used as a non-invasive clinical marker of intragraft endothelial cell injury.
Collapse
Affiliation(s)
- Manca Oblak
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Mlinšek
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nika Kojc
- Faculty of Medicine, Institute of Pathology, University of Ljubljana, Ljubljana, Slovenia
| | - Maja Frelih
- Faculty of Medicine, Institute of Pathology, University of Ljubljana, Ljubljana, Slovenia
| | - Jadranka Buturović-Ponikvar
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Arnol
- Department of Nephrology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
154
|
Lamarthée B, Burger C, Leclaire C, Lebraud E, Zablocki A, Morin L, Lebreton X, Charreau B, Snanoudj R, Charbonnier S, Blein T, Hardy M, Zuber J, Satchell S, Gallazzini M, Terzi F, Legendre C, Taupin JL, Rabant M, Tinel C, Anglicheau D. CRISPR/Cas9-Engineered HLA-Deleted Glomerular Endothelial Cells as a Tool to Predict Pathogenic Non-HLA Antibodies in Kidney Transplant Recipients. J Am Soc Nephrol 2021; 32:3231-3251. [PMID: 35167486 PMCID: PMC8638404 DOI: 10.1681/asn.2021050689] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/20/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND After kidney transplantation, donor-specific antibodies against human leukocyte antigen donor-specific antibodies (HLA-DSAs) drive antibody-mediated rejection (ABMR) and are associated with poor transplant outcomes. However, ABMR histology (ABMRh) is increasingly reported in kidney transplant recipients (KTRs) without HLA-DSAs, highlighting the emerging role of non-HLA antibodies (Abs). METHODS W e designed a non-HLA Ab detection immunoassay (NHADIA) using HLA class I and II-deficient glomerular endothelial cells (CiGEnCΔHLA) that had been previously generated through CRISPR/Cas9-induced B2M and CIITA gene disruption. Flow cytometry assessed the reactivity to non-HLA antigens of pretransplantation serum samples from 389 consecutive KTRs. The intensity of the signal observed with the NHADIA was associated with post-transplant graft histology assessed in 951 adequate biopsy specimens. RESULTS W e sequentially applied CRISPR/Cas9 to delete the B2M and CIITA genes to obtain a CiGEnCΔHLA clone. CiGEnCΔHLA cells remained indistinguishable from the parental cell line, CiGEnC, in terms of morphology and phenotype. Previous transplantation was the main determinant of the pretransplantation NHADIA result (P<0.001). Stratification of 3-month allograft biopsy specimens (n=298) according to pretransplantation NHADIA tertiles demonstrated that higher levels of non-HLA Abs positively correlated with increased glomerulitis (P=0.002), microvascular inflammation (P=0.003), and ABMRh (P=0.03). A pretransplantation NHADIA threshold of 1.87 strongly discriminated the KTRs with the highest risk of ABMRh (P=0.005, log-rank test). A multivariate Cox model confirmed that NHADIA status and HLA-DSAs were independent, yet synergistic, predictors of ABMRh. CONCLUSION The NHADIA identifies non-HLA Abs and strongly predicts graft endothelial injury independent of HLA-DSAs.
Collapse
Affiliation(s)
- Baptiste Lamarthée
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Carole Burger
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Charlotte Leclaire
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Emilie Lebraud
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Aniela Zablocki
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Lise Morin
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Xavier Lebreton
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Béatrice Charreau
- Center for Research in Transplantation and Immunology, INSERM UMR1064, IHU CESTI, LabEx IGO and LabEx Transplantex, Nantes University, Nantes, France
| | - Renaud Snanoudj
- Immunology and Histocompatibility Laboratory, Saint-Louis Hospital, AP-HP, LabEx Transplantex, INSERM U1160, University Paris Diderot, Paris, France
| | - Soëli Charbonnier
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM U1163, University of Paris, Paris, France
| | - Tifanie Blein
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM U1163, University of Paris, Paris, France
| | - Mélanie Hardy
- Immunology and Histocompatibility Laboratory, Saint-Louis Hospital, AP-HP, INSERM U976, IRSL, University of Paris, Paris, France
| | - Julien Zuber
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France,Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM U1163, University of Paris, Paris, France
| | - Simon Satchell
- Bristol Renal, Bristol Heart Institute, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Morgan Gallazzini
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Fabiola Terzi
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Christophe Legendre
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean Luc Taupin
- Immunology and Histocompatibility Laboratory, Saint-Louis Hospital, AP-HP, INSERM U976, IRSL, University of Paris, Paris, France
| | - Marion Rabant
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France,Department of Pathology, Necker Hospital, AP-HP, Paris, France
| | - Claire Tinel
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France
| | - Dany Anglicheau
- Necker-Enfants Malades Institute, Institut National de la Santé et de la Recherche Médicale (INSERM) U1151, University of Paris, Paris, France,Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| |
Collapse
|
155
|
Tatapudi VS, Kopchaliiska D, da Gente GJ, Buenaventura OF, Singh M, Laszik Z, Adey DB, Rajalingam R. Solid-Phase C1q/C3d Fixing Readouts Correlate with High Median Fluorescence Intensity (MFI) De Novo Donor-Specific HLA Antibodies and C4d⁺ Antibody-Mediated Rejection in Kidney Transplant Recipients. Ann Transplant 2021; 26:e934175. [PMID: 34848674 PMCID: PMC8647455 DOI: 10.12659/aot.934175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Solid-phase assays to investigate the complement-activating capacity of HLA antibodies have been utilized to optimize organ allocation and improve transplant outcomes. The clinical utility of C1q/C3d-binding characteristics of de novo donor-specific anti-HLA antibodies (dnDSA) associated with C4d-positive antibody-mediated rejection (C4d⁺ AMR) in kidney transplants (KTx) has not been defined. MATERIAL AND METHODS Sera from 120 KTx recipients that had dnDSA concurrent with protocol/cause biopsy (median 3.8 years after transplantation) were screened for C1q and C3d-binding dnDSA. The difference in the incidence of C4d⁺ AMR between recipients with and without C1q/C3d-binding dnDSA was assessed. RESULTS Over 86% of dnDSAs were class II antibodies. The immunodominant dnDSAs characterized by the highest median fluorescence intensity (MFI) in most recipients were HLA-DQ antibodies (67%). Most recipients (62%, n=74) had either C1q⁺ (56%), C3d⁺ (48%), or both C1q⁺C3d⁺ (41.2%) dnDSA, while the remaining 38% were negative for both C1q and C3d. Of those with C1q⁺/C3d⁺ dnDSA, 87% had high-MFI IgG (MFI=14144±5363 and 13932±5278, respectively), while 65% of C1q⁻C3d⁻ dnDSA had low-MFI IgG (MFI=5970±3347). The incidence of C4d+ AMR was significantly higher in recipients with C1q⁺ (66%), C3d+ (74%), and C1q⁺C3d⁺ (72%) dnDSA than in those with C1q⁻C3d⁻ dnDSA (30%) recipients. Recipients with C3d⁺/C1q⁺ dnDSA had higher C4d⁺ scores on biopsy. CONCLUSIONS C1q⁺/C3d⁺ dnDSA were associated with C4d⁺ AMR and high-IgG MFI. Our data call into question the predictive utility of C1q/C3d-binding assays in identifying KTx recipients at risk of allograft failure. In conclusion, IgG MFI is sufficient for clinical management, and the C1q/C3d-assays with added cost do not provide any additional information.
Collapse
Affiliation(s)
- Vasishta S Tatapudi
- Kidney Transplant Service, Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Dessislava Kopchaliiska
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Gilberto J da Gente
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Owen F Buenaventura
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Manpreet Singh
- Kidney Transplant Service, Department of Medicine,, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Zoltan Laszik
- Department of Pathology, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Deborah B Adey
- Kidney Transplant Service, Department of Medicine, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
156
|
Loupy A, Mengel M, Haas M. 30 years of the International Banff Classification for Allograft Pathology: The Past, Present and Future of Kidney Transplant Diagnostics. Kidney Int 2021; 101:678-691. [DOI: 10.1016/j.kint.2021.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/06/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
|
157
|
Taira H, Noguchi H, Ueki K, Kaku K, Tsuchimoto A, Okabe Y, Ohya Y, Nakamura M. Initiation of dialysis for kidney graft failure: A retrospective single-center cohort study. Ther Apher Dial 2021; 26:806-814. [PMID: 34779578 DOI: 10.1111/1744-9987.13756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/22/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022]
Abstract
Few studies have focused on the outcome of dialysis for kidney graft failure. We investigated the outcomes of dialysis for graft failure. We retrospectively studied 52 patients undergoing dialysis for graft failure at our facility from January 2004 to December 2018. The mean age at initiation of dialysis was 51.8 ± 13.5 years. The patient survival rates after initiation of dialysis at 1, 3, and 5 years were 96.0%, 93.8%, and 82.4%, respectively. The rate of unplanned initiation was 44.2%. In multivariate logistic analysis, lack of follow-up by nephrologists and pre-emptive kidney transplantation (PEKT) tended to be risk factors for unplanned initiation (P = 0.065 and P = 0.014, respectively). Our study suggests that the prognosis of patients with dialysis for graft failure is acceptable. Dialysis for graft failure, especially in patients with PEKT, tends to be unplanned, and for safe initiation, early involvement of nephrologists may be necessary.
Collapse
Affiliation(s)
- Hirona Taira
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Hiroshi Noguchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenji Ueki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keizo Kaku
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihiro Tsuchimoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuhiro Okabe
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Ohya
- Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
158
|
Del Bello A, Gaible C, Longlune N, Hebral AL, Esposito L, Gandia P, Kamar N. Tacrolimus Intrapatient Variability After Switching From Immediate or Prolonged-Release to Extended-Release Formulation, After an Organ Transplantation. Front Pharmacol 2021; 12:602764. [PMID: 34690747 PMCID: PMC8529208 DOI: 10.3389/fphar.2021.602764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 07/26/2021] [Indexed: 01/06/2023] Open
Abstract
Background and Purpose: Several formulations of tacrolimus are available, but evidence of the benefit of changing to the most recent formulations is lacking. Tacrolimus intra-patient variability (tacrolimus IPV) is an emerging risk factor associated with poor graft outcomes after solid organ transplantations. Here, we examined the modifications of tacrolimus IPV after switching to a different formulation of tacrolimus. Experimental Approach: We identified 353 solid organ transplant recipients that were switched in our center from immediate-release (IR-tacrolimus) or prolonged-release tacrolimus (PR-tacrolimus) to extended-release, LCP-tacrolimus (LCP-tacrolimus). Among them, 54 patients underwent at least 3 available tacrolimus blood concentrations before and after the switch, allowing us to investigate tacrolimus IPV. Key Results: The switch was considered as a safe procedure since only four of the 353 patients presented a graft rejection after the switch, and no patient was hospitalized for tacrolimus overdose. The tacrolimus IPV estimated by the coefficient of variation (CV-IPV) was stable before and after the switch to LCP-tacrolimus (CV-IPV: 29.0% (IQR 25-75 (15.5; 38.5) before and 24.0% (15.8; 36.5) after the switch, p = 0.65). Conclusion and Implications: Switching from IR- or PR-tacrolimus to LCP-tacrolimus is a safe procedure. However, the CV-tacrolimus IPV was not impacted by the change of formulation.
Collapse
Affiliation(s)
- Arnaud Del Bello
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France.,INSERM U1043, IFR-BMT, CHU Purpan, Toulouse, France.,Université Paul Sabatier, Toulouse, France
| | - Clotilde Gaible
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France
| | - Nathalie Longlune
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France
| | - Anne-Laure Hebral
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France
| | - Laure Esposito
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France
| | - Peggy Gandia
- Pharmacokinetics and Toxicology Laboratory, Toulouse University Hospital, Toulouse, France.,INTHERES, INRAE, ENVT, Université de Toulouse, Toulouse, France
| | - Nassim Kamar
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France.,INSERM U1043, IFR-BMT, CHU Purpan, Toulouse, France.,Université Paul Sabatier, Toulouse, France
| |
Collapse
|
159
|
Schrezenmeier E, Müller M, Friedersdorff F, Khadzhynov D, Halleck F, Staeck O, Dürr M, Zhang K, Eckardt KU, Budde K, Lehner LJ. Evaluation of severity of delayed graft function in kidney transplant recipients. Nephrol Dial Transplant 2021; 37:973-981. [PMID: 34665258 DOI: 10.1093/ndt/gfab304] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The most common definition of delayed graft function (DGF) relies on dialysis during the first week post-transplant and does not consider DGF severity. The impact of DGF severity on long-term graft outcome remains controversial. METHODS We analyzed 627 deceased-donor kidney transplant recipients (KTR) transplanted 2005-2015 at our center for DGF-severity, associated risk factors and long-term consequences of DGF. RESULTS We found 349 (55.7%) KTR with DGF, which were classified into 4 groups according to DGF duration (0-1, 2-7, 8-14, >14 days) and were compared to KTR with no DGF. Increasing duration of DGF was associated with progressive worsening of 10-year death-censored graft survival (no DGF: 88.3% (95%CI: 82.4-94.2), 0-1 day: 81.3% (95%CI: 68.2-94.4), 2-7 days: 61.5% (95%CI: 43.1.1-79.9), 8-14 days: 66.6% (95%CI: 47.4-85.8), >14 days: 51.2% (95%CI: 33-69.4); p < 0.001). In kidneys with a Kidney Donor Profile Index (KDPI) ≥85% all DGF severity groups demonstrated reduced graft survival. However, in < 85%KDPI kidneys, only > 14 days DGF duration showed worse outcomes. CONCLUSIONS DGF had a duration-dependent effect on graft survival, which varied depending on KDPI. Of note, 0-1-day DGF showed comparable results to no DGF in the whole cohort.
Collapse
Affiliation(s)
- Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Mia Müller
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Friedersdorff
- Department of Urology, Charité Universitätsmedizin Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Dmytro Khadzhynov
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver Staeck
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Dürr
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kun Zhang
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lukas J Lehner
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
160
|
Cornell LD. Histopathologic Features of Antibody Mediated Rejection: The Banff Classification and Beyond. Front Immunol 2021; 12:718122. [PMID: 34646262 PMCID: PMC8503253 DOI: 10.3389/fimmu.2021.718122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/07/2021] [Indexed: 01/27/2023] Open
Abstract
Antibody mediated rejection (ABMR) in the kidney can show a wide range of clinical presentations and histopathologic patterns. The Banff 2019 classification currently recognizes four diagnostic categories: 1. Active ABMR, 2. Chronic active ABMR, 3. Chronic (inactive) ABMR, and 4. C4d staining without evidence of rejection. This categorization is limited in that it does not adequately represent the spectrum of antibody associated injury in allograft, it is based on biopsy findings without incorporating clinical features (e.g., time post-transplant, de novo versus preformed DSA, protocol versus indication biopsy, complement inhibitor drugs), the scoring is not adequately reproducible, and the terminology is confusing. These limitations are particularly relevant in patients undergoing desensitization or positive crossmatch kidney transplantation. In this article, I discuss Banff criteria for these ABMR categories, with a focus on patients with pre-transplant DSA, and offer a framework for considering the continuum of allograft injury associated with donor specific antibody in these patients.
Collapse
Affiliation(s)
- Lynn D Cornell
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
161
|
Safety and Efficacy of Sodium-glucose Cotransporter 2 Inhibitors in Kidney Transplant Recipients With Pretransplant Type 2 Diabetes Mellitus: A Retrospective, Single-center, Inverse Probability of Treatment Weighting Analysis of 85 Transplant Patients. Transplant Direct 2021; 7:e772. [PMID: 34646935 PMCID: PMC8500668 DOI: 10.1097/txd.0000000000001228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/20/2021] [Accepted: 08/06/2021] [Indexed: 01/01/2023] Open
Abstract
Supplemental Digital Content is available in the text. Whether sodium-glucose cotransporter 2 (SGLT2) inhibitors can be used effectively and safely in kidney transplant (KT) recipients with pretransplant type 2 diabetes as the primary cause of end-stage renal disease (ESRD) remains unclear. In this study, we retrospectively analyzed the efficacy and safety of SGLT2 inhibitors compared with other oral hypoglycemic agents (OHAs) in KT recipients with pretransplant type 2 diabetes as the primary cause of ESRD.
Collapse
|
162
|
Caveolin-1 in Kidney Chronic Antibody-Mediated Rejection: An Integrated Immunohistochemical and Transcriptomic Analysis Based on the Banff Human Organ Transplant (B-HOT) Gene Panel. Biomedicines 2021; 9:biomedicines9101318. [PMID: 34680435 PMCID: PMC8533527 DOI: 10.3390/biomedicines9101318] [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: 08/16/2021] [Revised: 09/13/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023] Open
Abstract
Caveolin-1 overexpression has previously been reported as a marker of endothelial injury in kidney chronic antibody-mediated rejection (c-ABMR), but conclusive evidence supporting its use for daily diagnostic practice is missing. This study aims to evaluate if Caveolin-1 can be considered an immunohistochemical surrogate marker of c-ABMR. Caveolin-1 expression was analyzed in a selected series of 22 c-ABMR samples and 11 controls. Caveolin-1 immunohistochemistry proved positive in peritubular and glomerular capillaries of c-ABMR specimens, irrespective of C4d status whereas all controls were negative. Multiplex gene expression profiling in c-ABMR cases confirmed Caveolin-1 overexpression and identified additional genes (n = 220) and pathways, including MHC Class II antigen presentation and Type II interferon signaling. No differences in terms of gene expression (including Caveolin-1 gene) were observed according to C4d status. Conversely, immune cell signatures showed a NK-cell prevalence in C4d-negative samples compared with a B-cell predominance in C4d-positive cases, a finding confirmed by immunohistochemical assessment. Finally, differentially expressed genes were observed between c-ABMR and controls in pathways associated with Caveolin-1 functions (angiogenesis, cell metabolism and cell–ECM interaction). Based on our findings, Caveolin-1 resulted as a key player in c-ABMR, supporting its role as a marker of this condition irrespective of C4d status.
Collapse
|
163
|
Moktefi A, Hivelin M, Grimbert P, Carmagnat M, Sbidian E, Papouin B, Suberbielle C, Wolkenstein P, Bosc R, Meningaud JP, Lantieri L, Ortonne N. Face transplantation: A longitudinal histological study focusing on chronic active and mucosal rejection in a series with long-term follow-up. Am J Transplant 2021; 21:3088-3100. [PMID: 33445219 DOI: 10.1111/ajt.16489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 01/25/2023]
Abstract
The 2007 Banff working classification of skin-containing Tissue Allograft Pathology addressed only acute T cell-mediated rejection in skin. We report the longitudinal long-term histological follow-up of six face transplant recipients, focusing on chronic and mucosal rejection. We identified three patterns suggestive of chronic rejection (lichen planus-like, vitiligo-like and scleroderma-like). Four patients presented lichen planus-like and vitiligo-like chronic rejection at 52 ± 17 months posttransplant with severe concomitant acute T cell-mediated rejection. After lichen planus-like rejection, two patients developed scleroderma-like alterations. Graft vasculopathy with C4d deposits and de novo DSA led to subsequent graft loss in one patient. Chronic active rejection was frequent and similar patterns were noted in mucosae. Concordance between 124 paired skin and mucosal biopsies acute rejection grades was low (κ = 0.2, p = .005) but most grade 0/I mucosal rejections were associated with grade 0/I skin rejections. We defined discordant (grade≥II mucosal rejection and grade 0/I skin rejection) (n = 55 [70%]) and concordant (grade≥II rejection in both biopsies) groups. Mucosal biopsies of the discordant group displayed lower intra-epithelial GranzymeB/FoxP3 ratios suggesting a less aggressive phenotype (p = .08). The grading system for acute rejection in mucosa may require phenotyping. Whether discordant infiltrates reflect a latent allo-immune reaction leading to chronic rejection remains an open question.
Collapse
Affiliation(s)
- Anissa Moktefi
- APHP (Assistance Publique-Hôpitaux de Paris), Pathology Department, Groupe Hospitalier Henri-Mondor/Albert Chenevier, Créteil, France.,Univ Paris Est Creteil, Créteil, France
| | - Mikael Hivelin
- APHP (Assistance Publique-Hôpitaux de Paris), Department of Plastic Surgery, Hôpital Ambroise Paré, Boulogne, France.,Université Versailles Saint Quentin en Yvelines, Suresnes, France
| | - Philippe Grimbert
- Univ Paris Est Creteil, Créteil, France.,APHP (Assistance Publique-Hôpitaux de Paris), Nephrology and Renal Transplantation Department, Institut Francilien de Recherche en Néphrologie et Transplantation (IFRNT), Groupe Hospitalier Henri-Mondor/Albert-Chenevier, Créteil, France
| | - Maryvonnick Carmagnat
- APHP (Assistance Publique-Hôpitaux de Paris), Immunology and Histocompatibility Department, Saint Louis Hospital, Paris, France
| | - Emilie Sbidian
- Univ Paris Est Creteil, Créteil, France.,APHP (Assistance Publique-Hôpitaux de Paris), Department of Dermatology, Groupe Hospitalier Henri-Mondor/Albert Chenevier, Créteil, France
| | - Barbara Papouin
- APHP (Assistance Publique-Hôpitaux de Paris), Pathology Department, Groupe Hospitalier Henri-Mondor/Albert Chenevier, Créteil, France
| | - Caroline Suberbielle
- APHP (Assistance Publique-Hôpitaux de Paris), Immunology and Histocompatibility Department, Saint Louis Hospital, Paris, France.,Université Paris Diderot, Paris, France
| | - Pierre Wolkenstein
- APHP (Assistance Publique-Hôpitaux de Paris), Department of Dermatology, Groupe Hospitalier Henri-Mondor/Albert Chenevier, Créteil, France.,DHU (Département Hospitalo-Universitaire) VIC (Virus-Immunité-Cancer), Université Paris-Est-Créteil, (UPEC), IMRB (Institut Mondor de Recherche Biomédicale), Equipe Ortonne, INSERM U 955, Créteil, France
| | - Romain Bosc
- DHU (Département Hospitalo-Universitaire) VIC (Virus-Immunité-Cancer), Université Paris-Est-Créteil, (UPEC), IMRB (Institut Mondor de Recherche Biomédicale), Equipe Ortonne, INSERM U 955, Créteil, France.,APHP (Assistance Publique-Hôpitaux de Paris), Department of Plastic Surgery, Groupe Hospitalier Henri-Mondor/Albert Chenevier, Créteil, France
| | - Jean-Paul Meningaud
- DHU (Département Hospitalo-Universitaire) VIC (Virus-Immunité-Cancer), Université Paris-Est-Créteil, (UPEC), IMRB (Institut Mondor de Recherche Biomédicale), Equipe Ortonne, INSERM U 955, Créteil, France.,APHP (Assistance Publique-Hôpitaux de Paris), Department of Plastic Surgery, Groupe Hospitalier Henri-Mondor/Albert Chenevier, Créteil, France
| | - Laurent Lantieri
- APHP (Assistance Publique-Hôpitaux de Paris), Department of Plastic Surgery, Hôpital Européen Georges Pompidou (HEGP), Paris, France.,Université de Paris, Paris, France
| | - Nicolas Ortonne
- APHP (Assistance Publique-Hôpitaux de Paris), Pathology Department, Groupe Hospitalier Henri-Mondor/Albert Chenevier, Créteil, France.,DHU (Département Hospitalo-Universitaire) VIC (Virus-Immunité-Cancer), Université Paris-Est-Créteil, (UPEC), IMRB (Institut Mondor de Recherche Biomédicale), Equipe Ortonne, INSERM U 955, Créteil, France
| |
Collapse
|
164
|
Chancharoenthana W, Leelahavanichkul A, Ariyanon W, Vadcharavivad S, Phumratanaprapin W. Comparative Long-Term Renal Allograft Outcomes of Recurrent Immunoglobulin A with Severe Activity in Kidney Transplant Recipients with and without Rituximab: An Observational Cohort Study. J Clin Med 2021; 10:jcm10173939. [PMID: 34501386 PMCID: PMC8432075 DOI: 10.3390/jcm10173939] [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: 06/14/2021] [Revised: 08/09/2021] [Accepted: 08/28/2021] [Indexed: 12/17/2022] Open
Abstract
Recurrent IgA nephropathy (IgAN) remains an important cause of allograft loss in renal transplantation. Due to the limited efficacy of corticosteroid in the treatment of recurrent glomerulonephritis, rituximab was used in kidney transplant (KT) recipients with severe recurrent IgAN. A retrospective cohort study was conducted between January 2015 and December 2020. Accordingly, there were 64 KT recipients with biopsy-proven recurrent IgAN with similar baseline characteristics that were treated with the conventional standard therapy alone (controls, n = 43) or together with rituximab (cases, n = 21). All of the recipients had glomerular endocapillary hypercellularity and proteinuria (>1 g/d) with creatinine clearance (CrCl) > 30 mL/min/1.73 m2 and well-controlled blood pressure using renin–angiotensin–aldosterone blockers. The treatment outcomes were renal allograft survival rate, proteinuria, and post-treatment allograft pathology. During 3.8 years of follow-up, the rituximab-based regimen rapidly decreased proteinuria within 12 months after rituximab administration and maintained renal allograft function—the primary endpoint—for approximately 3 years. There were eight recipients in the case group (38%), and none in the control group reached a complete remission (proteinuria < 250 mg/d) at 12 months after treatment. Notably, renal allograft histopathology from patients with rituximab-based regimen showed the less severe endocapillary hypercellularity despite the remaining strong IgA deposition. In conclusion, adjunctive treatment with rituximab potentially demonstrated favorable outcomes for treatment of recurrent severe IgAN post-KT as demonstrated by proteinuria reduction and renal allograft function in our cohort. Further in-depth mechanistic studies with the longer follow-up periods are recommended.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Tropical Nephrology Research Unit, Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
- Correspondence: ; Tel.: +66-2256-4132; Fax: +66-2252-5952
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
- Translational Research in Inflammatory and Immunology Research Unit (TRIRU), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wassawon Ariyanon
- Cardiometabolic Centre, Department of Medicine, Bangkok Nursing Hospital, Bangkok 10500, Thailand;
| | - Somratai Vadcharavivad
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Weerapong Phumratanaprapin
- Tropical Nephrology Research Unit, Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand;
| |
Collapse
|
165
|
Kim SK, Yang JJ, Hwang SH, Sung H, Shin S, Ko SY, Oh HB. Prediction of HLA-DQ in Deceased Donors and its Clinical Significance in Kidney Transplantation. Ann Lab Med 2021; 41:190-197. [PMID: 33063680 PMCID: PMC7591288 DOI: 10.3343/alm.2021.41.2.190] [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: 04/06/2020] [Revised: 06/01/2020] [Accepted: 09/22/2020] [Indexed: 01/06/2023] Open
Abstract
Background HLA-DQ typing in deceased donors is not mandatory in Korea. Therefore, when patients develop DQ antibodies after kidney transplantation (KT) from deceased donor, it is impossible to determine whether they are donor-specific antibodies (DSA). We developed DQ prediction programs for the HLA gene and evaluated their clinical utility. Methods Two HLA-DQ prediction programs were developed: one based on Lewontin's linkage disequilibrium (LD) and haplotype frequency and the other on an artificial neural network (ANN). Low-resolution HLA-A, -B, -DR, and -DQ typing data of 5,603 Korean patients were analyzed in terms of haplotype frequency and used to develop an ANN DQ prediction program. Predicted DQ (pDQ) genotype accuracy was analyzed using the typed DQ data of 403 patients. pDQ DSA agreement, sensitivity, specificity, and false-negative rate was evaluated using 1,970 single-antigen bead assays performed on 885 KT recipients. The clinical significance of DQ and pDQ DSA was evaluated in 411 KT recipients. Results pDQ genotype accuracies were 75.4% (LD algorithm) and 75.7% (ANN). When the second most likely pDQ (LD algorithm) was also considered, the genotype accuracy increased to 92.6%. pDQ DSA (LD algorithm) agreement, sensitivity, specificity, and false-negative rate were 97.5%, 97.3%, 98.6%, and 2.4%, respectively. The antibody-mediated rejection treatment frequency was significantly higher in DQ or pDQ DSA-positive patients than in DQ or pDQ DSA-negative patients (P<0.001). Conclusions Our DQ prediction programs showed good accuracy and could aid DQ DSA detection in patients who had undergone deceased donor KT without donor HLA-DQ typing.
Collapse
Affiliation(s)
- Soo-Kyung Kim
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - John Jeongseok Yang
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Sang-Hyun Hwang
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Heungsup Sung
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Sung Shin
- Division of Kidney and Pancreas Transplantation, Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Sun-Young Ko
- Department of Laboratory Medicine, College of Medicine, Korea University, Seoul, Korea
| | - Heung-Bum Oh
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| |
Collapse
|
166
|
Cunha APL, Fabreti-Oliveira RA, Lasmar MF, Garcia JC, Vilela TP, Nascimento E. Clinical Outcome of Kidney Transplant Patients on the Allograft Function, Loss, Effects of HLA-DQB1-DSA +, and Graft Survival. Transplant Proc 2021; 53:2188-2196. [PMID: 34420780 DOI: 10.1016/j.transproceed.2021.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Matching for HLA-DQB1 molecules and anti-DQ donor-specific antibodies (DSAs) has been less studied to allocate transplants from deceased donors in developed countries. The aim of this study was to evaluate the clinical outcome of 519 kidney transplant recipients on the allograft function, loss, and survival and with emphasis on effects of HLA-DQB1-DSA+ at minimum of 10 years' follow-up. METHODS Five hundred nineteen kidney transplant patients were allocated into 3 groups (G) by immunologic profiles, namely, G1 (SPI-SAB HLA-DQ negative [DQ-]), G2 (SPI-SAB HLA-DQ positive DSA negative [DQ+/DSA-]), and G3 (SPI-SAB HLA-DQ DSA positive [DQ+ DSA+]), and the outcomes were reported until 10 years after transplantation. RESULTS The proportion of rejection episodes was higher in G3 (25.0% and 26.32%, respectively) than in G1 (8.63% and 6.82%, respectively) and G2 (10.0% and 0%, respectively; P = .047 and P = .014, respectively). In G3, 3 patients lost their grafts by antibody-mediated rejection. Patients who received kidneys from deceased donors (G3) showed worse graft survival rates than those from G1 donors (P = .001). Patients from G3 had a 2.18-fold higher risk of graft loss than patients from G1 (P = .028). CONCLUSION Allograft function was worse in G3 than in G2 or G1, and graft losses were more frequent by T-cell-mediated rejection in G1, and graft losses by antibody-mediated rejection were similar in G1 and G3 due to HLA class I (A1, 11 and B 8, 52) and HLA class II by DR7 and DQ 2, 5, 9 DSA, respectively. Allograft survival decreased in patients with HLA-DQB1 DSA. The risk of graft loss was 1.75-fold that in patients who received transplants from living donors.
Collapse
Affiliation(s)
- A P L Cunha
- University Hospital of the Faculty of Medical Science, Belo Horizonte, Minas Gerais, Brazil; Faculty of Medical Sciences, Belo Horizonte, Minas Gerais
| | - R A Fabreti-Oliveira
- University Hospital of the Faculty of Medical Science, Belo Horizonte, Minas Gerais, Brazil; IMUNOLAB-Laboratory of Histocompatibility, Belo Horizonte, Minas Gerais, Brazil
| | - M F Lasmar
- University Hospital of the Faculty of Medical Science, Belo Horizonte, Minas Gerais, Brazil; Faculty of Medical Sciences, Belo Horizonte, Minas Gerais
| | - J C Garcia
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais
| | - T P Vilela
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais
| | - E Nascimento
- IMUNOLAB-Laboratory of Histocompatibility, Belo Horizonte, Minas Gerais, Brazil; Institute of Education and Research of the Hospital Santa Casa, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
167
|
Parajuli S, Bath NM, Hidalgo L, Leverson G, Garg N, R Redfield R, Mandelbrot DA. Impact of low-level pretransplant donor-specific antibodies on outcomes after kidney transplantation. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1508-1519. [PMID: 34407300 PMCID: PMC8589373 DOI: 10.1002/iid3.504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/09/2021] [Accepted: 07/29/2021] [Indexed: 01/23/2023]
Abstract
Background The effect of low‐level pretransplant donor‐specific antibody (DSA) on kidney transplant outcomes is not well described. The goal of this study was to compare outcomes among patients of varying immunologic risk, based on the level of pretransplant DSA. Methods We retrospectively reviewed all adult kidney transplant recipients who had undergone a transplant at our center between January 2013 and May 2017. Patients were grouped as negative DSA (mean fluorescence intensity, [MFISUM < 100]), low‐level DSA (MFISUM 100–1000), and positive DSA (MFISUM > 1000). Rejection, infection, graft, and patient survival were outcomes measured. Results Of 952 patients, 82.1% had negative DSA, 10.7% had low‐level DSA, and 7.1% had positive DSA. The positive DSA group had the highest rate of antibody‐mediated rejection (10.3%), followed by low‐level DSA (7.8%) and the negative DSA group (4.5%) (p = .034). The rate of BK viremia was highest in the positive DSA group (39.7%), followed by the low‐level group (30.4%) and the negative DSA group (25.6%), (p = .025). None of the other outcomes, including graft or patient survival, were different between the groups. Conclusion While low‐level DSA should not prevent proceeding with kidney transplantation, it should not be ignored. Future studies are needed to investigate the long‐term effects of varying levels of pre‐transplant DSA on outcomes.
Collapse
Affiliation(s)
- Sandesh Parajuli
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Natalie M Bath
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Luis Hidalgo
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Glen Leverson
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Neetika Garg
- Division of Nephrology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Robert R Redfield
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.,Department of Surgery, Hospital of the University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Didier A Mandelbrot
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
168
|
Moris D, Cendales LC. Sensitization and Desensitization in Vascularized Composite Allotransplantation. Front Immunol 2021; 12:682180. [PMID: 34456906 PMCID: PMC8385557 DOI: 10.3389/fimmu.2021.682180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/27/2021] [Indexed: 01/02/2023] Open
Abstract
Vascularized composite allotransplantation (VCA) is a field under research and has emerged as an alternative option for the repair of severe disfiguring defects that result from severe tissue loss in a selected group of patients. Lifelong immunosuppressive therapy, immunosuppression associated complications, and the effects of the host immune response in the graft are major concerns in this type of quality-of-life transplant. The initial management of extensive soft tissue injury can lead to the development of anti-HLA antibodies through injury-related factors, transfusion and cadaveric grafting. The role of antibody-mediated rejection, donor-specific antibody (DSA) formation and graft rejection in the context of VCA still remain poorly understood. The most common antigenic target of preexisting alloantibodies are MHC mismatches, though recognition of ABO incompatible antigens, minor histocompatibility complexes and endothelial cells has also been shown to contribute to rejection. Mechanistically, alloantibody-mediated tissue damage occurs primarily through complement fixation as well as through antibody-dependent cellular toxicity. If DSA exist, activation of complement and coagulation cascades can result in vascular thrombosis and infarction and thus rejection and graft loss. Both preexisting DSA but especially de-novo DSA are currently considered as main contributors to late allograft injury and graft failure. Desensitization protocols are currently being developed for VCA, mainly including removal of alloantibodies whereas treatment of established antibody-mediated rejection is achieved through high dose intravenous immunoglobulins. The long-term efficacy of such therapies in sensitized VCA recipients is currently unknown. The current evidence base for sensitizing events and outcomes in reconstructive transplantation is limited. However, current data show that VCA transplantation has been performed in the setting of HLA-sensitization.
Collapse
Affiliation(s)
| | - Linda C. Cendales
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
169
|
Lee H, Park Y, Ban TH, Song SH, Song SH, Yang J, Ahn C, Yang CW, Chung BH. Synergistic impact of pre-sensitization and delayed graft function on allograft rejection in deceased donor kidney transplantation. Sci Rep 2021; 11:16095. [PMID: 34373479 PMCID: PMC8352860 DOI: 10.1038/s41598-021-95327-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/22/2021] [Indexed: 11/26/2022] Open
Abstract
The aim of this study is to investigate whether or not delayed graft function (DGF) and pre-transplant sensitization have synergistic adverse effects on allograft outcome after deceased donor kidney transplantation (DDKT) using the Korean Organ Transplantation Registry (KOTRY) database, the nationwide prospective cohort. The study included 1359 cases between May 2014 and June 2019. The cases were divided into 4 subgroups according to pre-sensitization and the development of DGF post-transplant [non-pre-sensitized-DGF(−) (n = 1097), non-pre-sensitized-DGF(+) (n = 127), pre-sensitized-DGF(−) (n = 116), and pre-sensitized-DGF(+) (n = 19)]. We compared the incidence of biopsy-proven allograft rejection (BPAR), time-related change in allograft function, allograft or patient survival, and post-transplant complications across 4 subgroups. The incidence of acute antibody-mediated rejection (ABMR) was significantly higher in the pre-sensitized-DGF(+) subgroup than in other 3 subgroups. In addition, multivariable cox regression analysis demonstrated that pre-sensitization combined with DGF is an independent risk factor for the development of acute ABMR (hazard ratio 4.855, 95% confidence interval 1.499–15.727). Moreover, DGF and pre-sensitization showed significant interaction (p-value for interaction = 0.008). Pre-sensitization combined with DGF did not show significant impact on allograft function, and allograft or patient survival. In conclusion, the combination of pre-sensitization and DGF showed significant synergistic interaction on the development of allograft rejection after DDKT.
Collapse
Affiliation(s)
- Hanbi Lee
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea
| | - Yohan Park
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea.,Division of Nephrology, Department of Internal Medicine, Konyang University Hospital, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Tae Hyun Ban
- Division of Nephrology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, Seoul, South Korea
| | - Sang Heon Song
- Organ Transplantation Center and Department of Internal Medicine, Pusan National University Hospital, Busan, South Korea
| | - Seung Hwan Song
- Department of Surgery, Ewha Womans University Medical Center, Seoul, South Korea
| | - Jaeseok Yang
- Department of Nephrology, Seoul National University Hospital, Seoul, South Korea
| | - Curie Ahn
- Department of Nephrology, Seoul National University Hospital, Seoul, South Korea
| | - Chul Woo Yang
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea
| | - Byung Ha Chung
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea.
| | | |
Collapse
|
170
|
Hooper DK, Varnell CD, Rich K, Carle A, Huber J, Mostajabi F, Dahale D, Pai ALH, Goebel J, Modi AC. A Medication Adherence Promotion System to Reduce Late Kidney Allograft Rejection: A Quality Improvement Study. Am J Kidney Dis 2021; 79:335-346. [PMID: 34352285 DOI: 10.1053/j.ajkd.2021.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/06/2021] [Indexed: 11/11/2022]
Abstract
RATIONALE AND OBJECTIVE Adolescent and young adult kidney transplant recipients have high risk of rejection related to suboptimal adherence. Multi-component interventions improve adherence in controlled trials, but clinical implementation is lacking. We describe an initiative to reduce allograft rejection using evidence-based adherence promotion clinical strategies. STUDY DESIGN Interrupted time series. SETTING AND PARTICIPANTS Kidney transplant recipients cared for at Cincinnati Children's Hospital ≥1 year post-transplant and taking ≥1 immunosuppressive medication from 2014 through 2017. QUALITY IMPROVEMENT ACTIVITIES The following interventions were implemented over 14 months: 1) adherence promotion training for clinical staff, 2) EHR-supported adherence risk screening, 3) systematic assessment of medication adherence barriers, 4) designation of specific staff to address adherence barriers, 5) shared decision-making with the patients to overcome adherence barriers, 6) follow-up to assess progress, 7) optional electronic medication monitoring. OUTCOMES Primary Outcome: Late acute rejection. Process measures: barriers assessments performed, barriers identified, number of interventions performed. Secondary outcomes/balancing measures: de novo DSA, biopsy rate, rejections per biopsy. ANALYTICAL APPROACH Time series analysis using statistical process control, evaluated patient-days between acute rejections as well as monthly rejections per 100 patient-months before and after implementation. To control for known rejection risk factors including changes in treatment and case mix, multivariable analyses were performed. RESULTS The monthly rejection rate fell from 1.61 rejections/100 patient-months in the 26-months pre-implementation to 0.88 rejections/100 patient-months in the 22-months post-implementation. In multivariable analysis, MAPS was associated with a 50% reduction in rejection incidence (IRR 0.50, 95% CI: 0.27-0.91, p=0.02). DSA and time since transplant were also associated with rejection incidence (IRR 2.27, p=0.02 and IRR 0.87, p=0.02, respectively) LIMITATIONS: A single center study. Potential confounding by unmeasured variables. CONCLUSIONS Clinical implementation of evidence-based adherence-promotion strategies was associated with a 50% reduction in acute rejection incidence over two years.
Collapse
Affiliation(s)
- David K Hooper
- Division of Nephrology (MLC-7022), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229; University of Cincinnati, College of Medicine, CARE/Crawley Building Suite E-870, 3230 Eden Avenue, PO BOX 670555, Cincinnati, OH 45267-0555; James M Anderson Center for Health Systems Excellence, (MLC-7014), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229.
| | - Charles D Varnell
- Division of Nephrology (MLC-7022), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229; University of Cincinnati, College of Medicine, CARE/Crawley Building Suite E-870, 3230 Eden Avenue, PO BOX 670555, Cincinnati, OH 45267-0555; James M Anderson Center for Health Systems Excellence, (MLC-7014), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229
| | - Kristin Rich
- University of Cincinnati, College of Medicine, CARE/Crawley Building Suite E-870, 3230 Eden Avenue, PO BOX 670555, Cincinnati, OH 45267-0555; Division of Behavioral Medicine and Clinical Psychology (MLC-3015), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229
| | - Adam Carle
- University of Cincinnati, College of Medicine, CARE/Crawley Building Suite E-870, 3230 Eden Avenue, PO BOX 670555, Cincinnati, OH 45267-0555; James M Anderson Center for Health Systems Excellence, (MLC-7014), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229; Univerisity of Cincinnati, Collage of Arts and Sciences, Department of Psychology, 155 B McMicken Hall Cincinnati, OH 45221
| | - John Huber
- Department of Information Services (MLC 9009), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229
| | - Farida Mostajabi
- James M Anderson Center for Health Systems Excellence, (MLC-7014), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229
| | - Devesh Dahale
- Southeast Alabama Medical Center, 1108 Ross Clark Circle, Dothan, AL 36301
| | - Ahna L H Pai
- University of Cincinnati, College of Medicine, CARE/Crawley Building Suite E-870, 3230 Eden Avenue, PO BOX 670555, Cincinnati, OH 45267-0555; Division of Behavioral Medicine and Clinical Psychology (MLC-3015), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229
| | - Jens Goebel
- Section of Pediatric Nephrology, Helen DeVos Children's Hospital, 100 Michigan Street NE, Grand Rapids, MI 49503; Department of Pediatrics and Human Development, Michigan State University, Life Sciences Bldg. 1355 Bogue St., B240, East Lansing MI 48824
| | - Avani C Modi
- University of Cincinnati, College of Medicine, CARE/Crawley Building Suite E-870, 3230 Eden Avenue, PO BOX 670555, Cincinnati, OH 45267-0555; Division of Behavioral Medicine and Clinical Psychology (MLC-3015), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave. Cincinnati, OH 45229
| |
Collapse
|
171
|
Varnell C, Harshman LA, Smith L, Liu C, Chen S, Al‐Akash S, Barletta G, Belsha C, Brakeman P, Chaudhuri A, Fadakar P, Garro R, Gluck C, Goebel J, Kershaw D, Matossian D, Nailescu C, Patel HP, Pruette C, Ranabothu S, Rodig N, Smith J, Sebestyen VanSickle J, Weng P, Danziger‐Isakov L, Hooper DK, Seifert M. COVID‐19 in pediatric kidney transplantation: The Improving Renal Outcomes Collaborative. Am J Transplant 2021. [DOI: 10.1111/ajt.16501 10.1111/ajt.16501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Charles Varnell
- Cincinnati Children’s Hospital Medical Center Cincinnati Ohio
- University of Cincinnati College of Medicine Cincinnati Ohio
| | | | - Laurie Smith
- Cincinnati Children’s Hospital Medical Center Cincinnati Ohio
| | - Chunyan Liu
- Cincinnati Children’s Hospital Medical Center Cincinnati Ohio
| | - Shiran Chen
- Cincinnati Children’s Hospital Medical Center Cincinnati Ohio
| | | | | | - Craig Belsha
- SSM Health Cardinal Glennon Children's Hospital Saint Louis Missouri
| | - Paul Brakeman
- Department of Pediatrics University of California San Francisco California
| | - Abanti Chaudhuri
- Lucile Packard Children’s HospitalStanford University Stanford California
| | - Paul Fadakar
- UPMC Children’s Hospital of Pittsburgh Pittsburgh Pennsylvania
| | - Rouba Garro
- Children’s Healthcare of AtlantaEmory School of Medicine Atlanta Georgia
| | - Caroline Gluck
- Nemours/Alfred I. duPont Hospital for Children Wilmington Delaware
| | - Jens Goebel
- Children’s Hospital Colorado Aurora Colorado
| | | | - Debora Matossian
- Ann & Robert H. Lurie Children’s Hospital of Chicago Chicago Illinois
| | - Corina Nailescu
- Riley Hospital for Children at Indiana University Health Indianapolis Indiana
| | | | - Cozumel Pruette
- Johns Hopkins University School of Medicine Baltimore Maryland
| | | | - Nancy Rodig
- Boston Children’s HospitalHarvard Medical School Boston Massachusetts
| | - Jodi Smith
- Seattle Children’s Hospital Seattle Washington
| | | | - Patricia Weng
- UCLA Mattel Children’s Hospital Los Angeles California
| | - Lara Danziger‐Isakov
- Cincinnati Children’s Hospital Medical Center Cincinnati Ohio
- University of Cincinnati College of Medicine Cincinnati Ohio
| | - David K. Hooper
- Cincinnati Children’s Hospital Medical Center Cincinnati Ohio
- University of Cincinnati College of Medicine Cincinnati Ohio
| | - Michael Seifert
- University of Alabama at BirminghamChildren’s of Alabama Birmingham Alabama
| |
Collapse
|
172
|
Varnell CD, Rich KL, Zhang B, Carle AC, Pai ALH, Modi AC, Hooper DK. Predicting acute rejection in children, adolescents, and young adults with a kidney transplant by assessing barriers to taking medication. Pediatr Nephrol 2021; 36:2453-2461. [PMID: 33501558 PMCID: PMC8263481 DOI: 10.1007/s00467-021-04946-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/15/2020] [Accepted: 01/12/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND Nonadherence to immunosuppression is common among pediatric, adolescent, and young adult kidney transplant recipients and a leading cause of graft loss. Assessing barriers to medication adherence in clinical practice may identify patients at risk for rejection and provide therapeutic targets. METHODS Kidney transplant patients and/or their caregivers were assessed for 14 barriers to medication adherence using the barriers assessment tool. We compared rejection rates between patients with at least one reported adherence barrier to those without reported adherence barriers using a Kaplan-Meier estimator and Cox proportional hazard models to adjust for other mediators of acute rejection at 2 years following barriers assessment. RESULTS Ninety-eight patients were assessed for barriers to adherence. Over the 2-year observation period, 22 patients developed biopsy-proven acute rejection (BPAR). Kaplan-Meier estimates show that patients with an identified barrier to adherence were more likely to have BPAR (p = 0.02) than patients without an identified barrier in the 24 months following barriers assessment. The median time to rejection for patients who experienced acute rejection was 175.5 days (IQR 63-276 days) from the time of barriers assessment. An identified barrier to adherence remained the only statistically significant predictor of BPAR with Cox modeling (HR 2.6, p = 0.04), after accounting for age, sex, and race. CONCLUSIONS Pediatric and adolescent kidney transplant recipients with identified adherence barriers are at increased risk for acute rejection. Barriers to adherence provide a potentially modifiable therapeutic target that can be assessed in clinic to guide targeted interventions.
Collapse
Affiliation(s)
- Charles D Varnell
- Division of Nephrology & Hypertension, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, MLC 7022, Cincinnati, OH, 45229, USA. .,James M. Anderson Center for Health Systems Excellence, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Kristin L Rich
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.,Division of Behavioral and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bin Zhang
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.,Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adam C Carle
- James M. Anderson Center for Health Systems Excellence, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Psychology, College of Arts and Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Ahna L H Pai
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.,Division of Behavioral and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Avani C Modi
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.,Division of Behavioral and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David K Hooper
- Division of Nephrology & Hypertension, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, MLC 7022, Cincinnati, OH, 45229, USA.,James M. Anderson Center for Health Systems Excellence, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
173
|
McCaffrey J, Shenoy M. Acute rejection and growth outcomes in paediatric kidney allograft recipients treated with a corticosteroid minimisation immunosuppressive protocol. Pediatr Nephrol 2021; 36:2463-2472. [PMID: 33560455 DOI: 10.1007/s00467-021-04948-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/16/2020] [Accepted: 01/12/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Corticosteroid minimisation immunosuppressive protocols (CMP) for children are an approach to safely reduce unwanted medication side effects associated with long-term exposure following kidney transplantation. Here, we provide data regarding the incidence of acute rejection and growth over an extended follow-up in children receiving the CMP used in our centre. METHODS We retrospectively analysed all children treated with a CMP who received a kidney transplant and had follow-up care in our centre between 2009 and 2019. Data were compared to 5 control groups from recent studies. RESULTS Ninety-nine kidney allograft recipients were included in the study (mean follow-up 4.4 years). There was no difference in the cumulative frequency of acute rejection in CMP-treated graft recipients compared to controls. Graft function at latest follow-up was significantly lower in graft recipients experiencing acute rejection compared to those without acute rejection (53.7 mL/min/1.73 m2 vs. 66.8 mL/min/1.73 m2, p = 0.021). Children experiencing >1 acute rejection episode had a greatly elevated risk of graft failure (p = 0.0009, OR 68.25). At latest follow-up, 64/90 (71.1%) graft recipients had a normal height, and younger graft recipients demonstrated greater catch up growth than older children. CMP-treated graft recipients showed a reduced rate of height deficit (28.9% vs. 55.1%, p = 0.0025), less obesity (12.2% vs. 23.9%, p = 0.031), and reduced rates of hypertension (35.4% vs. 68.2%, p< 0.0001). CONCLUSIONS Children treated with a CMP show greater height attainment, lower frequency of obesity, and reduced rates of hypertension, without an increased risk of acute rejection. Graphical abstract.
Collapse
Affiliation(s)
- James McCaffrey
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| | - Mohan Shenoy
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
174
|
Varnell Jr C, Harshman LA, Smith L, Liu C, Chen S, Al-Akash S, Barletta GM, Belsha C, Brakeman P, Chaudhuri A, Fadakar P, Garro R, Gluck C, Goebel J, Kershaw D, Matossian D, Nailescu C, Patel HP, Pruette C, Ranabothu S, Rodig N, Smith J, Sebestyen VanSickle J, Weng P, Danziger-Isakov L, Hooper DK, Seifert M. COVID-19 in pediatric kidney transplantation: The Improving Renal Outcomes Collaborative. Am J Transplant 2021; 21:2740-2748. [PMID: 33452854 PMCID: PMC8013281 DOI: 10.1111/ajt.16501] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/17/2020] [Accepted: 01/10/2021] [Indexed: 01/25/2023]
Abstract
There are limited data on the impact of COVID-19 in children with a kidney transplant (KT). We conducted a prospective cohort study through the Improving Renal Outcomes Collaborative (IROC) to collect clinical outcome data about COVID-19 in pediatric KT patients. Twenty-two IROC centers that care for 2732 patients submitted testing and outcomes data for 281 patients tested for SARS-CoV-2 by PCR. Testing indications included symptoms and/or potential exposures to COVID-19 (N = 134, 47.7%) and/or testing per hospital policy (N = 154, 54.8%). Overall, 24 (8.5%) patients tested positive, of which 15 (63%) were symptomatic. Of the COVID-19-positive patients, 16 were managed as outpatients, six received non-ICU inpatient care and two were admitted to the ICU. There were no episodes of respiratory failure, allograft loss, or death associated with COVID-19. To estimate incidence, subanalysis was performed for 13 centers that care for 1686 patients that submitted all negative and positive COVID-19 results. Of the 229 tested patients at these 13 centers, 10 (5 asymptomatic) patients tested positive, yielding an overall incidence of 0.6% and an incidence among tested patients of 4.4%. Pediatric KT patients in the United States had a low estimated incidence of COVID-19 disease and excellent short-term outcomes.
Collapse
Affiliation(s)
- Charles Varnell Jr
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Laurie Smith
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Chunyan Liu
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Shiran Chen
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | | | | | - Craig Belsha
- SSM Health Cardinal Glennon Children’s Hospital, Saint Louis, Missouri
| | - Paul Brakeman
- Department of Pediatrics, University of California, San Francisco, California
| | - Abanti Chaudhuri
- Lucile Packard Children’s Hospital, Stanford University, Stanford, California
| | - Paul Fadakar
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rouba Garro
- Children’s Healthcare of Atlanta, Emory School of Medicine, Atlanta, Georgia
| | - Caroline Gluck
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, Delaware
| | - Jens Goebel
- Children’s Hospital Colorado, Aurora, Colorado
- Present address Jens Goebel, Helen DeVos Children’s Hospital, Grand Rapids, MichiganMichigan, USA
| | | | - Debora Matossian
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Corina Nailescu
- Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana
| | | | - Cozumel Pruette
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Nancy Rodig
- Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jodi Smith
- Seattle Children’s Hospital, Seattle, Washington
| | | | - Patricia Weng
- UCLA Mattel Children’s Hospital, Los Angeles, California
| | - Lara Danziger-Isakov
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David K. Hooper
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael Seifert
- University of Alabama at Birmingham, Children’s of Alabama, Birmingham, Alabama
| |
Collapse
|
175
|
Multiplex gene analysis reveals T-cell and antibody-mediated rejection-specific upregulation of complement in renal transplants. Sci Rep 2021; 11:15464. [PMID: 34326417 PMCID: PMC8322413 DOI: 10.1038/s41598-021-94954-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/13/2021] [Indexed: 01/03/2023] Open
Abstract
In renal transplantation, complement is involved in ischemia reperfusion injury, graft rejection and dysfunction. However, it is still unclear how induction of complement and its activation are initiated. Using allograft biopsies of a well-characterized cohort of 28 renal transplant patients with no rejection (Ctrl), delayed graft function (DGF), acute T-cell-mediated (TCMR) or antibody-mediated rejection (ABMR) we analyzed differences in complement reaction. For that mRNA was isolated from FFPE sections, quantified with a multiplex gene expression panel and correlated with transplant conditions and follow-up of patients. Additionally, inflammatory cells were quantified by multiplex immunohistochemistry. In allograft biopsies with TCMR and ABMR gene expression of C1QB was 2-4 fold elevated compared to Ctrl. In TCMR biopsies, mRNA counts of several complement-related genes including C1S, C3, CFB and complement regulators CFH, CR1 and SERPING1 were significantly increased compared to Ctrl. Interestingly, expression levels of about 75% of the analyzed complement related genes correlated with cold ischemia time (CIT) and markers of inflammation. In conclusion, this study suggest an important role of complement in transplant pathology which seems to be at least in part triggered by CIT. Multiplex mRNA analysis might be a useful method to refine diagnosis and explore new pathways involved in rejection.
Collapse
|
176
|
|
177
|
Lehner LJ, Öllinger R, Globke B, Naik MG, Budde K, Pratschke J, Eckardt KU, Kahl A, Zhang K, Halleck F. Impact of Early Pancreatic Graft Loss on Outcome after Simultaneous Pancreas-Kidney Transplantation (SPKT)-A Landmark Analysis. J Clin Med 2021; 10:jcm10153237. [PMID: 34362019 PMCID: PMC8347953 DOI: 10.3390/jcm10153237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 01/06/2023] Open
Abstract
(1) Background: Simultaneous pancreas-kidney transplantation (SPKT) is a standard therapeutic option for patients with diabetes mellitus type I and kidney failure. Early pancreas allograft failure is a complication potentially associated with worse outcomes. (2) Methods: We performed a landmark analysis to assess the impact of early pancreas graft loss within 3 months on mortality and kidney graft survival over 10 years. This retrospective single-center study included 114 adult patients who underwent an SPKT between 2005 and 2018. (3) Results: Pancreas graft survival rate was 85.1% at 3 months. The main causes of early pancreas graft loss were thrombosis (6.1%), necrosis (2.6%), and pancreatitis (2.6%). Early pancreas graft loss was not associated with reduced patient survival (p = 0.168) or major adverse cerebral or cardiovascular events over 10 years (p = 0.741) compared to patients with functioning pancreas, after 3 months. Moreover, kidney graft function (p = 0.494) and survival (p = 0.461) were not significantly influenced by early pancreas graft loss. (4) Conclusion: In this study, using the landmark analysis technique, early pancreas graft loss within 3 months did not significantly impact patient or kidney graft survival over 10 years.
Collapse
Affiliation(s)
- Lukas Johannes Lehner
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.G.N.); (K.B.); (K.-U.E.); (A.K.); (F.H.)
- Correspondence: ; Tel.: +49-30-45-051-4002
| | - Robert Öllinger
- Department of Surgery, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (R.Ö.); (B.G.); (J.P.)
| | - Brigitta Globke
- Department of Surgery, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (R.Ö.); (B.G.); (J.P.)
| | - Marcel G. Naik
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.G.N.); (K.B.); (K.-U.E.); (A.K.); (F.H.)
- Berlin Institute of Health (BIH), 10117 Berlin, Germany;
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.G.N.); (K.B.); (K.-U.E.); (A.K.); (F.H.)
| | - Johann Pratschke
- Department of Surgery, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (R.Ö.); (B.G.); (J.P.)
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.G.N.); (K.B.); (K.-U.E.); (A.K.); (F.H.)
| | - Andreas Kahl
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.G.N.); (K.B.); (K.-U.E.); (A.K.); (F.H.)
| | - Kun Zhang
- Berlin Institute of Health (BIH), 10117 Berlin, Germany;
- Department of Internal Medicine and Cardiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany; (M.G.N.); (K.B.); (K.-U.E.); (A.K.); (F.H.)
| |
Collapse
|
178
|
Goesch TR, Wilson NA, Zeng W, Verhoven BM, Zhong W, Coumbe Gitter MM, Fahl WE. Suppression of Inflammation-Associated Kidney Damage Post-Transplant Using the New PrC-210 Free Radical Scavenger in Rats. Biomolecules 2021; 11:1054. [PMID: 34356678 PMCID: PMC8301928 DOI: 10.3390/biom11071054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/12/2021] [Accepted: 07/12/2021] [Indexed: 01/10/2023] Open
Abstract
Allograft kidney transplantation, which triggers host cellular- and antibody-mediated rejection of the kidney, is a major contributor to kidney damage during transplant. Here, we asked whether PrC-210 would suppress damage seen in allograft kidney transplant. Brown Norway (BN) rat kidneys were perfused in situ (UW Solution) with or without added 30 mM PrC-210, and then immediately transplanted into Lewis (LEW) rats. 20 h later, the transplanted BN kidneys and LEW rat plasma were analyzed. Kidney histology, and kidney/serum levels of several inflammation-associated cytokines, were measured to assess mismatch-related kidney pathology, and PrC-210 protective efficacy. Twenty hours after the allograft transplants: (i) significant histologic kidney tubule damage and mononuclear inflammatory cell infiltration were seen in allograft kidneys; (ii) kidney function metrics (creatinine and BUN) were significantly elevated; (iii) significant changes in key cytokines, i.e., TIMP-1, TNF-alpha and MIP-3A/CCL20, and kidney activated caspase levels were seen. In PrC-210-treated kidneys and recipient rats, (i) kidney histologic damage (Banff Scores) and mononuclear infiltration were reduced to untreated background levels; (ii) creatinine and BUN were significantly reduced; and (iii) activated caspase and cytokine changes were significantly reduced, some to background. In conclusion, the results suggest that PrC-210 could provide broadly applicable organ protection for many allograft transplantation conditions; it could protect transplanted kidneys during and after all stages of the transplantation process-from organ donation, through transportation, re-implantation and the post-operative inflammation-to minimize acute and chronic rejection.
Collapse
Affiliation(s)
| | - Nancy A. Wilson
- Department of Surgery, Division of Organ Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.W.); (W.Z.); (B.M.V.); (W.Z.)
| | - Weifeng Zeng
- Department of Surgery, Division of Organ Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.W.); (W.Z.); (B.M.V.); (W.Z.)
| | - Bret M. Verhoven
- Department of Surgery, Division of Organ Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.W.); (W.Z.); (B.M.V.); (W.Z.)
| | - Weixiong Zhong
- Department of Surgery, Division of Organ Transplant, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.W.); (W.Z.); (B.M.V.); (W.Z.)
| | - Maya M. Coumbe Gitter
- Department of Oncology, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - William E. Fahl
- Obvia Pharmaceuticals Ltd., Madison, WI 53719, USA;
- Department of Oncology, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI 53706, USA;
| |
Collapse
|
179
|
HLA Antibody Incompatible Renal Transplantation: Long-term Outcomes Similar to Deceased Donor Transplantation. Transplant Direct 2021; 7:e732. [PMID: 34291154 PMCID: PMC8291351 DOI: 10.1097/txd.0000000000001183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 11/14/2022] Open
Abstract
Background. HLA incompatible renal transplantation still remains one of best therapeutic options for a subgroup of patients who are highly sensitized and difficult to match but not much is known about its long-term graft and patient survival. Methods. One hundred thirty-four HLA incompatible renal transplantation patients from 2003 to 2018 with a median follow of 6.93 y were analyzed retrospectively to estimate patient and graft survivals. Outcomes were compared with groups defined by baseline crossmatch status and the type and timings of rejection episodes. Results. The overall patient survival was 95%, 90%, and 81%; and graft survival was 95%, 85%, and 70% at 1, 5, and 10 y, respectively. This was similar to the first-time deceased donor transplant cohort. The graft survival for pretreatment cytotoxic-dependent crossmatch (CDC) positive crossmatch group was significantly low at 83%, 64%, and 40% at 1, 5, and 10 y, respectively, compared with other groups (Bead/CDC, P = 0.007; CDC/Flow, P = 0.001; and microbead assay/flow cytometry crossmatch, P = 0.837), although those with a low CDC titer (<1 in 2) have comparable outcomes to the CDC negative group. Female patients in general fared worse in both patient and graft survival outcomes in each of the 3 groups based on pretreatment crossmatch, although this did not reach statistical significance. Antibody-mediated rejection was the most frequent type of rejection with significant decline in graft survival by 10 y when compared with no rejection (P < 0.001). Rejection that occurred or continued to occur after the first 2 wk of transplantation caused a significant reduction in graft survivals (P < 0.001), whereas good outcomes were seen in those with a single early rejection episode. Conclusions. One-, 5-, and 10-y HLA incompatible graft and patient survival is comparable to deceased donor transplantation and can be further improved by excluding high-CDC titer cases. Antibody-positive female patients show worse long-term survival. Resolution of early rejection is associated with good long-term graft survival.
Collapse
|
180
|
Cherukuri A, Salama AD, Mehta R, Mohib K, Zheng L, Magee C, Harber M, Stauss H, Baker RJ, Tevar A, Landsittel D, Lakkis FG, Hariharan S, Rothstein DM. Transitional B cell cytokines predict renal allograft outcomes. Sci Transl Med 2021; 13:13/582/eabe4929. [PMID: 33627487 DOI: 10.1126/scitranslmed.abe4929] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
Early immunological biomarkers that predict rejection and chronic allograft loss are needed to inform preemptive therapy and improve long-term outcomes. Here, we prospectively examined the ratio of interleukin-10 (IL-10) to tumor necrosis factor-α (TNFα) produced by transitional-1 B cells (T1B) 3 months after transplantation as a predictive biomarker for clinical and subclinical renal allograft rejection and subsequent clinical course. In both Training (n = 162) and Internal Validation (n = 82) Sets, the T1B IL-10/TNFα ratio 3 months after transplantation predicted both clinical and subclinical rejection anytime in the first year. The biomarker also predicted subsequent late rejection with a lead time averaging 8 months. Among biomarker high-risk patients, 60% had early rejection, of which 48% recurred later in the first posttransplant year. Among high-risk patients without early rejection, 74% developed rejection later in the first year. In contrast, only 5% of low-risk patients had early and 5% late rejection. The biomarker also predicted rejection in an External Validation Set (n = 95) and in key patient subgroups, confirming generalizability. Biomarker high-risk patients exhibited progressively worse renal function and decreased 5-year graft survival compared to low-risk patients. Treatment of B cells with anti-TNFα in vitro augmented the IL-10/TNFα ratio, restored regulatory activity, and inhibited plasmablast differentiation. To conclude, the T1B IL-10/TNFα ratio was validated as a strong predictive biomarker of renal allograft outcomes and provides a rationale for preemptive therapeutic intervention with TNF blockade.
Collapse
Affiliation(s)
- Aravind Cherukuri
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Alan D Salama
- University College of London Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Rajil Mehta
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Kanishka Mohib
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Leting Zheng
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Rheumatology and Immunology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ciara Magee
- University College of London Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Mark Harber
- University College of London Department of Renal Medicine, Royal Free Hospital, London NW3 2QG, UK
| | - Hans Stauss
- UCL Institute of Immunity and Transplantation, Royal Free Hospital, London NW3 2QG, UK
| | - Richard J Baker
- Renal Unit, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Amit Tevar
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Douglas Landsittel
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - Fadi G Lakkis
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sundaram Hariharan
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David M Rothstein
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA. .,Renal and Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
181
|
Cone BD, Zhang JQ, Sosa RA, Calabrese F, Reed EF, Fishbein GA. Phosphorylated S6 ribosomal protein expression by immunohistochemistry correlates with de novo donor-specific HLA antibodies in lung allograft recipients. J Heart Lung Transplant 2021; 40:1164-1171. [PMID: 34330604 DOI: 10.1016/j.healun.2021.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 06/12/2021] [Accepted: 06/28/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Per the ISHLT 2016 definition, a C4d-positive lung biopsy is required to meet criteria for definite antibody-mediated rejection (AMR). Unfortunately, C4d has poor sensitivity and specificity, and low inter-rater reliability. Phosphorylated S6 ribosomal protein (p-S6RP) expressed via the mTOR pathway has been shown to be a biomarker of AMR and correlates with donor-specific antibodies (DSA) in heart allografts. However, p-S6RP immunohistochemistry (IHC) in the setting of pulmonary AMR has yet to be evaluated. We sought to determine whether p-S6RP IHC performed on lung biopsies correlates with de novo DSA. METHODS IHC for p-S6RP performed on 26 biopsies from lung transplant recipients with de novo HLA DSA (DSA+) and 28 biopsies from patients with no DSA (DSA-) were evaluated by 3 pathologists who independently scored the degree of alveolar macrophage and pneumocyte staining. Staining in ≥50% of the biopsy as determined by at least 2 pathologists was considered positive. RESULTS Twenty-one (81%) DSA+ biopsies stained positive for p-S6RP in pneumocytes and 21 (81%) in macrophages. Six DSA- biopsies (21%) stained positive for p-S6RP in pneumocytes, 6 (21%) were positive in macrophages. Pneumocyte p-S6RP staining was 81% sensitive and 79% specific for DSA. Macrophage staining showed the same sensitivity and specificity but with lower inter-rater agreement (κ = 0.53 vs 0.68). CONCLUSIONS This study demonstrates a positive relationship between de novo DSA and p-S6RP expression in pneumocytes and macrophages using IHC. p-S6RP is relatively sensitive and specific, and has superior inter-rater reliability compared to C4d.
Collapse
Affiliation(s)
- Brian D Cone
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jennifer Q Zhang
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Rebecca A Sosa
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Elaine F Reed
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | |
Collapse
|
182
|
Miyairi S, Ueda D, Yagisawa T, Okada D, Keslar KS, Tanabe K, Dvorina N, Valujskikh A, Baldwin WM, Hazen SL, Fairchild RL. Recipient myeloperoxidase-producing cells regulate antibody-mediated acute versus chronic kidney allograft rejection. JCI Insight 2021; 6:148747. [PMID: 34081629 PMCID: PMC8410093 DOI: 10.1172/jci.insight.148747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022] Open
Abstract
Antibody-mediated rejection (ABMR) continues to be a major problem undermining the success of kidney transplantation. Acute ABMR of kidney grafts is characterized by neutrophil and monocyte margination in the tubular capillaries and by graft transcripts indicating NK cell activation, but the myeloid cell mechanisms required for acute ABMR have remained unclear. Dysregulated donor-specific antibody (DSA) responses with high antibody titers are induced in B6.CCR5-/- mice transplanted with complete MHC-mismatched A/J kidneys and are required for rejection of the grafts. This study tested the role of recipient myeloid cell production of myeloperoxidase (MPO) in the cellular and molecular components of acute ABMR. Despite induction of equivalent DSA titers, B6.CCR5-/- recipients rejected A/J kidneys between days 18 and 25, with acute ABMR, whereas B6.CCR5-/-MPO-/- recipients rejected the grafts between days 46 and 54, with histopathological features of chronic graft injury. On day 15, myeloid cells infiltrating grafts from B6.CCR5-/- and B6.CCR5-/-MPO-/- recipients expressed marked phenotypic and functional transcript differences that correlated with the development of acute versus chronic allograft injury, respectively. Near the time of peak DSA titers, activation of NK cells to proliferate and express CD107a was decreased within allografts in B6.CCR5-/-MPO-/- recipients. Despite high titers of DSA, depletion of neutrophils reproduced the inhibition of NK cell activation and decreased macrophage infiltration but increased monocytes producing MPO. Overall, recipient myeloid cells producing MPO regulate graft-infiltrating monocyte/macrophage function and NK cell activation that are required for DSA-mediated acute kidney allograft injury, and their absence switches DSA-mediated acute pathology and graft outcomes to chronic ABMR.
Collapse
Affiliation(s)
- Satoshi Miyairi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Urology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Daisuke Ueda
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Takafumi Yagisawa
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Urology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Daigo Okada
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Urology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Karen S. Keslar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - William M. Baldwin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stanley L. Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Robert L. Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
183
|
Crespo M, Llinàs-Mallol L, Redondo-Pachón D, Butler C, Gimeno J, Pérez-Sáez MJ, Burballa C, Buxeda A, Arias-Cabrales C, Folgueiras M, Sanz-Ureña S, Valenzuela NM, Reed EF, Pascual J. Non-HLA Antibodies and Epitope Mismatches in Kidney Transplant Recipients With Histological Antibody-Mediated Rejection. Front Immunol 2021; 12:703457. [PMID: 34305943 PMCID: PMC8300190 DOI: 10.3389/fimmu.2021.703457] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/15/2021] [Indexed: 01/03/2023] Open
Abstract
Background Correlation between antibody-mediated rejection (ABMR) and circulating HLA donor-specific antibodies (HLA-DSA) is strong but imperfect in kidney transplant (KT) recipients, raising the possibility of undetected HLA-DSA or non-HLA antibodies contributing to ABMR. Detailed evaluation of the degree of HLA matching together with the identification of non-HLA antibodies in KT may help to decipher the antibody involved. Methods We retrospectively assessed patients with transplant biopsies scored following Banff'15 classification. Pre- and post-transplant serum samples were checked for HLA and non-HLA antibodies [MICA-Ab, angiotensin-II type-1-receptor (AT1R)-Ab, endothelin-1 type-A-receptor (ETAR)-Ab and crossmatches with primary aortic endothelial cells (EC-XM)]. We also analyzed HLA epitope mismatches (HLA-EM) between donors and recipients to explore their role in ABMR histology (ABMRh) with and without HLA-DSA. Results One-hundred eighteen patients with normal histology (n = 19), ABMRh (n = 52) or IFTA (n = 47) were studied. ABMRh patients were HLA-DSApos (n = 38, 73%) or HLA-DSAneg (n = 14, 27%). Pre-transplant HLA-DSA and AT1R-Ab were more frequent in ABMRh compared with IFTA and normal histology cases (p = 0.006 and 0.003), without differences in other non-HLA antibodies. Only three ABMRhDSAneg cases showed non-HLA antibodies. ABMRhDSAneg and ABMRhDSApos cases showed similar biopsy changes and graft-survival. Both total class II and DRB1 HLA-EM were associated with ABMRhDSApos but not with ABMRhDSAneg. Multivariate analysis showed that pre-transplant HLA-DSA (OR: 3.69 [1.31-10.37], p = 0.013) and AT1R-Ab (OR: 5.47 [1.78-16.76], p = 0.003) were independent predictors of ABMRhDSApos. Conclusions In conclusion, pre-transplant AT1R-Ab is frequently found in ABMRhDSApos patients. However, AT1R-Ab, MICA-Ab, ETAR-Ab or EC-XM+ are rarely found among ABMRhDSAneg patients. Pre-transplant AT1R-Ab may act synergistically with preformed or de novo HLA-DSA to produce ABMRhDSApos but not ABMRhDSAneg. HLA epitope mismatch associates with ABMRhDSApos compared with ABMRhDSAneg, suggesting factors other than HLA are responsible for the damage.
Collapse
Affiliation(s)
- Marta Crespo
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Laura Llinàs-Mallol
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Dolores Redondo-Pachón
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Carrie Butler
- UCLA Immunogenetics Center, University of California Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Javier Gimeno
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - María José Pérez-Sáez
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Carla Burballa
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Anna Buxeda
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Carlos Arias-Cabrales
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Montserrat Folgueiras
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Sara Sanz-Ureña
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Nicole M. Valenzuela
- UCLA Immunogenetics Center, University of California Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Elaine F. Reed
- UCLA Immunogenetics Center, University of California Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
184
|
Rodelo J, González LA, Ustáriz J, Matera S, Pérez K, Ramírez Z, Arias LF, García Á, Arbeláez M, Henao J. Kidney transplantation outcomes in lupus nephritis: A 37-year single-center experience from Latin America. Lupus 2021; 30:1644-1659. [PMID: 34225520 DOI: 10.1177/09612033211028663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE We assessed patient and graft outcomes and prognostic factors in kidney transplantation in patients with end-stage kidney disease (ESKD) secondary to lupus nephritis (LN) undergoing kidney transplantation from August 1977 to December 2014 in a Latin American single center. METHODS The primary endpoint was patient survival, and the secondary endpoints were death-censored graft survival for the first renal transplant and the rate of recurrent LN (RLN). Kaplan-Meier method was used for survival analysis. Factors predicting patient and death-censored graft survivals were examined by Cox proportional-hazards regression analyses. RESULTS 185 patients were retrospectively evaluated. Patient survival rates were 88% at one year, 82% at three years, 78% at five years, and 67% at ten years. Death-censored graft survival for the first renal transplant was 93% at one year, 89% at three years, 87% at five years, and 80% at ten years. RLN was diagnosed in 2 patients (1.08%), but no graft was lost because of RLN. Thirty-nine (21.1%) patients died, and 65 (35.1%) patients experienced graft loss during the follow-up. By multivariable analyses, older recipient age and 1-month posttransplantation eGFR <45 ml/min/1.73m2 were associated with lower patient survival and an increased risk of graft loss, while induction immunosuppressive therapy exerted a protective effect on patients' survival. In the subgroup of patients in whom disease activity was measured at the time of transplantation, a higher SLEDAI score was also associated with lower patient survival and an increased risk of graft loss. CONCLUSION In a mostly Mestizo population, kidney transplantation is an excellent therapeutic alternative in LN patients with ESKD. Older recipient age, an eGFR <45 ml/min/1.73m2 at one month posttransplantation, and disease activity at the time of transplantation are predictive of a lower patient and death-censored graft survival, while induction immunosuppressive therapy has a protective effect on patient survival. RLN is rare and does not influence the risk of graft loss.
Collapse
Affiliation(s)
- Joaquín Rodelo
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Luis Alonso González
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - José Ustáriz
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Silvia Matera
- Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Keylis Pérez
- Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Zoraida Ramírez
- Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Luis Fernando Arias
- Department of Pathology, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Álvaro García
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia.,Division of Nephrology, Nefron Sas, Medellín, Colombia
| | - Mario Arbeláez
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia.,Division of Nephrology, Nefron Sas, Medellín, Colombia
| | - Jorge Henao
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Universidad de Antioquia, 27983Universidad de Antioquia, Hospital Universitario San Vicente Fundación, Medellín, Colombia.,Division of Nephrology, Nefron Sas, Medellín, Colombia
| |
Collapse
|
185
|
Leino AD, Park JM, Pasternak AL. Impact of CYP3A5 phenotype on tacrolimus time in therapeutic range and clinical outcomes in pediatric renal and heart transplant recipients. Pharmacotherapy 2021; 41:649-657. [PMID: 34129685 DOI: 10.1002/phar.2601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/11/2022]
Abstract
STUDY OBJECTIVE This study investigated the effect of CYP3A5 phenotype on time in therapeutic range (TTR) of tacrolimus post-transplant in pediatric patients. DESIGN AND DATA SOURCE This retrospective study assessed medical records of pediatric kidney and heart recipients with available CYP3A5 genotype for tacrolimus dosing, troughs, and the clinical events (biopsy-proven acute rejection [BPAR] and de novo donor-specific antibodies [dnDSA]). MEASUREMENTS AND MAIN RESULTS The primary outcome, mean TTR in the first 90 days post-transplant, was 9.0% (95% CI: -16.1, -1.9) lower in CYP3A5 expressers (p = 0.014) when adjusting for time to therapeutic concentration and organ type. There was no difference between CYP3A5 phenotypes in time to the first clinical event using TTR during the first 90 days. When applying TTR over the first year, there was a significant difference in event-free survival (EFS) which was 50.0% for CYP3A5 expressers/TTR < 35%, 45.5% for expressers/TTR ≥ 35%, 38.1% for nonexpressers/TTR < 35%, and 72.9% for nonexpressers/TTR ≥ 35% (log-rank p = 0.03). A post hoc analysis of EFS identified CYP3A5 expressers had lower EFS compared to nonexpressers in patients with TTR ≥ 35% (p = 0.04) but no difference among patients with TTR < 35% (p = 0.6). CONCLUSIONS The relationship between TTR and CYP3A5 phenotype suggests that achieving a TTR ≥ 35% during the first year may be a modifiable factor to attenuate the risk of BPAR and dnDSA.
Collapse
Affiliation(s)
- Abbie D Leino
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Jeong M Park
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy L Pasternak
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
186
|
Callemeyn J, Ameye H, Lerut E, Senev A, Coemans M, Van Loon E, Sprangers B, Van Sandt V, Rabeyrin M, Dubois V, Thaunat O, Kuypers D, Emonds MP, Naesens M. Revisiting the changes in the Banff classification for antibody-mediated rejection after kidney transplantation. Am J Transplant 2021; 21:2413-2423. [PMID: 33382185 DOI: 10.1111/ajt.16474] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 01/25/2023]
Abstract
The Banff classification for antibody-mediated rejection (ABMR) has undergone important changes, mainly by inclusion of C4d-negative ABMR in Banff'13 and elimination of suspicious ABMR (sABMR) with the use of C4d as surrogate for HLA-DSA in Banff'17. We aimed to evaluate the numerical and prognostic repercussions of these changes in a single-center cohort study of 949 single kidney transplantations, comprising 3662 biopsies that were classified according to the different versions of the Banff classification. Overall, the number of ABMR and sABMR cases increased from Banff'01 to Banff'13. In Banff'17, 248 of 292 sABMR biopsies were reclassified to No ABMR, and 44 of 292 to ABMR. However, reclassified sABMR biopsies had worse and better outcome than No ABMR and ABMR, which was mainly driven by the presence of microvascular inflammation and absence of HLA-DSA, respectively. Consequently, the discriminative performance for allograft failure was lowest in Banff'17, and highest in Banff'13. Our data suggest that the clinical and histological heterogeneity of ABMR is inadequately represented in a binary classification system. This study provides a framework to evaluate the updates of the Banff classification and assess the impact of proposed changes on the number of cases and risk stratification. Two alternative classifications introducing an intermediate category are explored.
Collapse
Affiliation(s)
- Jasper Callemeyn
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Heleen Ameye
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Evelyne Lerut
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Aleksandar Senev
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.,Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Maarten Coemans
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Elisabet Van Loon
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Ben Sprangers
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Vicky Van Sandt
- Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Maud Rabeyrin
- Department of Pathology, Hospices Civils de Lyon, Bron, France
| | - Valérie Dubois
- French National Blood Service (EFS), HLA Laboratory, Décines-Charpieu, France
| | - Olivier Thaunat
- Medical Research (Inserm) Unit 111, French National Institute of Health, Lyon, France.,Department of Transplantation, Nephrology and Clinical Immunology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Dirk Kuypers
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Marie-Paule Emonds
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.,Histocompatibility and Immunogenetics Laboratory, Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Maarten Naesens
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| |
Collapse
|
187
|
Schinstock CA, Askar M, Bagnasco SM, Batal I, Bow L, Budde K, Campbell P, Carroll R, Clahsen-van Groningen MC, Cooper M, Cornell LD, Cozzi E, Dadhania D, Diekmann F, Hesselink DA, Jackson AM, Kikic Z, Lower F, Naesens M, Roelofs JJ, Sapir-Pichhadze R, Kraus ES. A 2020 Banff Antibody-mediatedInjury Working Group examination of international practices for diagnosing antibody-mediated rejection in kidney transplantation - a cohort study. Transpl Int 2021; 34:488-498. [PMID: 33423340 DOI: 10.1111/tri.13813] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/24/2020] [Accepted: 01/02/2021] [Indexed: 12/24/2022]
Abstract
The Banff antibody-mediated rejection (ABMR) classification is vulnerable to misinterpretation, but the reasons are unclear. To better understand this vulnerability, we evaluated how ABMR is diagnosed in practice. To do this, the Banff Antibody-Mediated Injury Workgroup electronically surveyed an international cohort of nephrologists/surgeons (n = 133) and renal pathologists (n = 99). Most providers (97%) responded that they use the Banff ABMR classification at least sometimes, but DSA information is often not readily available. Only 41.1% (55/133) of nephrologists/surgeons and 19.2% (19/99) of pathologists reported that they always have DSA results when the biopsy is available. Additionally, only 19.6% (26/133) of nephrologists/surgeons responded that non-HLA antibody or molecular transcripts are obtained when ABMR histologic features are present but DSA is undetected. Several respondents agreed that histologic features concerning for ABMR in the absence of DSA and/or C4d are not well accounted for in the current classification [31.3% (31/99) pathologists and 37.6% (50/133) nephrologist/surgeons]. The Banff ABMR classification appears widely accepted, but efforts to improve the accessibility of DSA information for the multidisciplinary care team are needed. Further clarity is also needed in Banff ABMR nomenclature to account for the spectrum of ABMR and for histologic features suspicious for ABMR when DSA is absent.
Collapse
Affiliation(s)
- Carrie A Schinstock
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, USA
| | - Medhat Askar
- Baylor University Medical Center, Dallas, TX, USA.,Texas A&M Health Science Center Collect of Medicine, Bryan, TX, USA
| | - Serena M Bagnasco
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ibrahim Batal
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Laurine Bow
- Department of Transplantation Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Klemens Budde
- Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Patricia Campbell
- Department of Medicine and Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Robert Carroll
- Transplantation Immunogenetics Service, Australian Red Cross Blood Service Melbourne, Melbourne, Vic., Australia.,University of South Australia, Adelaide, SA, Australia
| | | | - Matthew Cooper
- Medstar Georgetown Transplant Institute, Washington, DC, USA
| | - Lynn D Cornell
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Emanuele Cozzi
- Transplant Immunology Unit, Department of Cardiac, Thoracic and Vascular Sciences, Padua University Hospital, Padua, Italy
| | - Darshana Dadhania
- Department of Medicine, Weill Cornell Medicine - New York Presbyterian Hospital, New York, NY, USA
| | - Fritz Diekmann
- Kidney Transplant Unit, Institut d'Incestigacions Biomèdiques August Pi i Sunyer, Hospital Clínic, Barcelona, Spain
| | - Dennis A Hesselink
- Department of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Zeljko Kikic
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Fritz Lower
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| | - Maarten Naesens
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium.,Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Joris J Roelofs
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ruth Sapir-Pichhadze
- Centre for Outcomes Research & Evaluation Research Institute, McGill University Health Center, Montreal, QC, Canada
| | - Edward S Kraus
- Division of Nephrology/Transplant Nephrology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
188
|
Kläger J, Eskandary F, Böhmig GA, Kozakowski N, Kainz A, Colin Aronovicz Y, Cartron JP, Segerer S, Regele H. Renal allograft DARCness in subclinical acute and chronic active ABMR. Transpl Int 2021; 34:1494-1505. [PMID: 33983671 PMCID: PMC8453966 DOI: 10.1111/tri.13904] [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: 01/10/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/13/2023]
Abstract
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.
Collapse
Affiliation(s)
- Johannes Kläger
- Department of Pathology, Medical University Vienna, Vienna, Austria
| | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | | | - Alexander Kainz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Yves Colin Aronovicz
- Laboratoire d'Excellence GR-Ex, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France
| | - Jean-Pierre Cartron
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm U1134, Institut National de la Transfusion Sanguine, Unité Biologie Intégrée du Globule Rouge, Paris, France
| | - Stephan Segerer
- Division of Nephrology, Dialysis and Transplantation, Kantonsspital Aarau, Aarau, Switzerland
| | - Heinz Regele
- Department of Pathology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
189
|
Yu MY, Kwon S, Moon JJ, Kim YC, Song EY, Lee H, Moon KC, Ha J, Kim DK, Han SW, Kim GH, Kim YS, Yang SH. Role of the IL-33/ST2 pathway in renal allograft rejection. Exp Cell Res 2021; 405:112705. [PMID: 34166678 DOI: 10.1016/j.yexcr.2021.112705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 11/25/2022]
Abstract
The interleukin-33 (IL-33)/suppression of tumorigenicity 2 (ST2) pathway modulates immune response and inflammation, associated with allograft dysfunction and rejection. We hypothesized that IL-33/ST2 is a marker of renal allograft rejection and IL-33/ST2 expression may differ according to rejection type. IL-33/ST2 expression was measured in sera and kidney tissues from recipients with acute antibody-mediated rejection (AAMR), acute cell-mediated rejection (ACMR), chronic antibody-mediated rejection (CAMR), and healthy controls. The soluble ST2 and IL-33/ST2 expression levels were higher in participants with all three rejection types than in controls. Although the expression levels in recipients with AAMR and ACMR were significantly higher than those with CAMR, there was no significant difference between the expression levels in AAMR and ACMR. Although IL-33, IL-8, and fibronectin expression were significantly increased after the addition of the recipients' serum in primary cultured human renal proximal tubular epithelial cells, the levels decreased after treatment with an anti-ST2 antibody. Furthermore, the anti-ST2 antibody specifically suppressed the upregulation of the mixed lymphocyte reaction. Boyden chamber assays demonstrated that anti-ST2 antibody abrogated chemotaxis induced by recombinant IL-33. Thus, IL-33 and ST2 are potent mediators of rejection. Treatment with an anti-ST2 antibody ameliorates rejection and could be a potential therapeutic strategy for renal allograft rejection.
Collapse
Affiliation(s)
- Mi-Yeon Yu
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea; Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Soie Kwon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Jong Joo Moon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yong-Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Eun Young Song
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyung Chul Moon
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Jongwon Ha
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang-Woong Han
- Department of Internal Medicine, Hanyang University Guri Hospital, Guri, South Korea; Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Gheun-Ho Kim
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, South Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, South Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|
190
|
Yamashita M, Lin MY, Hou J, Ren KY, Haas M. The Continuing Need for Electron Microscopy in Examination of Medical Renal Biopsies: Examples in Practice. GLOMERULAR DISEASES 2021; 1:145-159. [PMID: 36751496 PMCID: PMC9677721 DOI: 10.1159/000516831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/23/2021] [Indexed: 12/24/2022]
Abstract
Background For the better part of the past 6 decades, transmission electron microscopy (EM), together with routine light microscopy and immunofluorescence and/or immunohistochemistry (IHC), has been an essential component of the diagnostic workup of medical renal biopsies, particularly native renal biopsies, with increasing frequency in renal allograft biopsies as well. Studies performed prior to the year 2000 have indeed shown that a substantial fraction of renal biopsies cannot be accurately diagnosed without EM. Still, EM remains costly and labor-intensive, and with increasing pressure to reduce healthcare costs, some centers are de-emphasizing diagnostic EM. This trend has been coupled with advances in IHC and other methods in renal biopsy diagnosis over the past 2-3 decades. Summary Nonetheless, it has been our experience that the diagnostic value of EM in the comprehensive evaluation of renal biopsies remains similar to what it was 20-30 years ago. In this review, we provide several key examples from our practice where EM was essential in making the correct renal biopsy diagnosis, ranging from relatively common glomerular lesions to rare diseases. Key Messages EM remains an important component of the diagnostic evaluation of medical renal biopsies. Failure to perform EM in certain cases will result in an incorrect diagnosis, with possible clinical consequences. We strongly recommend that tissue for EM be taken and stored in an appropriate fixative and ultrastructural studies be performed for all native renal biopsies, as well as appropriate renal allograft biopsies as recommended by the Banff consortium.
Collapse
|
191
|
Kuscu C, Kiran M, Mohammed A, Kuscu C, Satpathy S, Wolen A, Bardhi E, Bajwa A, Eason JD, Maluf D, Mas V, Akalin E. Integrative Analyses of Circulating Small RNAs and Kidney Graft Transcriptome in Transplant Glomerulopathy. Int J Mol Sci 2021; 22:ijms22126218. [PMID: 34207555 PMCID: PMC8226568 DOI: 10.3390/ijms22126218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/01/2021] [Accepted: 06/06/2021] [Indexed: 02/07/2023] Open
Abstract
Transplant glomerulopathy develops through multiple mechanisms, including donor-specific antibodies, T cells and innate immunity. This study investigates circulating small RNA profiles in serum samples of kidney transplant recipients with biopsy-proven transplant glomerulopathy. Among total small RNA population, miRNAs were the most abundant species in the serum of kidney transplant patients. In addition, fragments arising from mature tRNA and rRNA were detected. Most of the tRNA fragments were generated from 5′ ends of mature tRNA and mainly from two parental tRNAs: tRNA-Gly and tRNA-Glu. Moreover, transplant patients with transplant glomerulopathy displayed a novel tRNA fragments signature. Gene expression analysis from allograft tissues demonstrated changes in canonical pathways related to immune activation such as iCos-iCosL signaling pathway in T helper cells, Th1 and Th2 activation pathway, and dendritic cell maturation. mRNA targets of down-regulated miRNAs such as miR-1224-5p, miR-4508, miR-320, miR-378a from serum were globally upregulated in tissue. Integration of serum miRNA profiles with tissue gene expression showed that changes in serum miRNAs support the role of T-cell mediated mechanisms in ongoing allograft injury.
Collapse
Affiliation(s)
- Canan Kuscu
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.K.); (A.W.); (A.B.); (J.D.E.)
- Correspondence: ; Tel.: +1-901-448-3162
| | - Manjari Kiran
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India; (M.K.); (S.S.)
| | - Akram Mohammed
- Center for Biomedical Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Cem Kuscu
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.K.); (A.W.); (A.B.); (J.D.E.)
| | - Sarthak Satpathy
- Department of Systems and Computational Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India; (M.K.); (S.S.)
| | - Aaron Wolen
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.K.); (A.W.); (A.B.); (J.D.E.)
| | - Elissa Bardhi
- Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (E.B.); (D.M.); (V.M.)
| | - Amandeep Bajwa
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.K.); (A.W.); (A.B.); (J.D.E.)
| | - James D. Eason
- Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, School of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.K.); (A.W.); (A.B.); (J.D.E.)
| | - Daniel Maluf
- Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (E.B.); (D.M.); (V.M.)
| | - Valeria Mas
- Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (E.B.); (D.M.); (V.M.)
| | - Enver Akalin
- Montefiore Medical Center, Abdominal Transplant Program, Albert Einstein College of Medicine, Bronx, NY 10467, USA;
| |
Collapse
|
192
|
Impact of Subclinical and Clinical Kidney Allograft Rejection Within 1 Year Posttransplantation Among Compatible Transplant With Steroid Withdrawal Protocol. Transplant Direct 2021; 7:e706. [PMID: 34124342 PMCID: PMC8191698 DOI: 10.1097/txd.0000000000001132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 12/31/2022] Open
Abstract
Early acute kidney rejection remains an important clinical issue. Methods The current study included 552 recipients who had 1-2 surveillance or indication biopsy within the 1 y posttransplant. We evaluated the impact of type of allograft inflammation on allograft outcome. They were divided into 5 groups: no inflammation (NI: 95), subclinical inflammation (SCI: 244), subclinical T cell-mediated rejection (TCMR) (SC-TCMR: 110), clinical TCMR (C-TCMR: 83), and antibody-mediated rejection (AMR: 20). Estimated glomerular filtration rate (eGFR) over time using linear mixed model, cumulative chronic allograft scores/interstitial fibrosis and tubular atrophy (IFTA) ≥2 at 12 mo, and survival estimates were compared between groups. Results The common types of rejections were C-TCMR (15%), SC-TCMR (19.9%), and AMR (3.6%) of patients. Eighteen of 20 patients with AMR had mixed rejection with TCMR. Key findings were as follows: (i) posttransplant renal function: eGFR was lower for patients with C-TCMR and AMR (P < 0.0001) compared with NI, SCI, and SC-TCMR groups. There was an increase in delta-creatinine from 3 to 12 mo and cumulative allograft chronicity scores at 12 mo (P < 0.001) according to the type of allograft inflammation. (ii) Allograft histology: the odds of IFTA ≥2 was higher for SC-TCMR (3.7 [1.3-10.4]; P = 0.04) but was not significant for C-TCMR (3.1 [1.0-9.4]; P = 0.26), and AMR (2.5 [0.5-12.8]; P = 0.84) compared with NI group, and (iii) graft loss: C-TCMR accounted for the largest number of graft losses and impending graft losses on long-term follow-up. Graft loss among patient with AMR was numerically higher but was not statistically significant. Conclusions The type of kidney allograft inflammation predicted posttransplant eGFR, cumulative chronic allograft score/IFTA ≥2 at 12 mo, and graft loss.
Collapse
|
193
|
Akdogan N, Ersoy-Evans S, Gokoz O, Erdem Y, Nasir S. Early recognition of chronic rejection in a face allotransplant patient with alopecia. J Cutan Pathol 2021; 48:1286-1297. [PMID: 34085296 DOI: 10.1111/cup.14069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/06/2021] [Accepted: 05/30/2021] [Indexed: 11/26/2022]
Abstract
The features of chronic rejection (CR) in full-face vascularized composite allotransplantation (VCA) are not well-known. Herein, we report a full-face transplant patient that experienced two episodes of acute rejection (AR) and three episodes of AR/CR over the course of 6-years. The patient noticed a small, round patch of hair loss in his beard 9 months after the second AR episode, which occurred 21 months post-transplantation. Biopsy of the alopecic patch showed lichen-planopilaris-like features, which was suggestive of early CR. Despite an increase in immunosuppressive dosages, the alopecia progressed. Following the second and third AR/CR episodes, the alopecia became more pronounced, with the addition of hyperpigmentation as well as sclerosis and telangiectasia. The findings of multiple biopsies showed CR. Based on these findings we think that alopecia with lichen-planopilaris-like histopathological features similar to grade III AR features, particularly in hair follicles appears to be an early finding of CR in the presented patient. The findings further indicate that follicular involvement may be a significant feature of CR in VCA patients and that it can present prior to sclerosis, vasculopathy, or loss of adnexa. The present case is uniquely important because of the distinctive presentation of CR, with hair follicles clinically and histopathologically affected, leading to progressive and irreversible alopecia with lichen-planopilaris-like histopathology.
Collapse
Affiliation(s)
- Neslihan Akdogan
- Department of Dermatology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Sibel Ersoy-Evans
- Department of Dermatology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Ozay Gokoz
- Department of Pathology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Yunus Erdem
- Department of Nephrology, School of Medicine, Hacettepe University, Ankara, Turkey
| | - Serdar Nasir
- Department of Plastic, Reconstructive and Aesthetic Surgery, School of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
194
|
Manook M, Flores WJ, Schmitz R, Fitch Z, Yoon J, Bae Y, Shaw B, Kirk A, Harnois M, Permar S, Farris AB, Magnani DM, Kwun J, Knechtle S. Measuring the Impact of Targeting FcRn-Mediated IgG Recycling on Donor-Specific Alloantibodies in a Sensitized NHP Model. Front Immunol 2021; 12:660900. [PMID: 34149698 PMCID: PMC8207189 DOI: 10.3389/fimmu.2021.660900] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/17/2021] [Indexed: 11/13/2022] Open
Abstract
Background In transplantation, plasmapheresis and IVIg provide the mainstay of treatment directed at reducing or removing circulating donor-specific antibody (DSA), yet both have limitations. We sought to test the efficacy of targeting the IgG recycling mechanism of the neonatal Fc receptor (FcRn) using anti-FcRn mAb therapy in a sensitized non-human primate (NHP) model, as a pharmacological means of lowering DSA. Methods Six (6) rhesus macaque monkeys, previously sensitized by skin transplantation, received a single dose of 30mg/kg anti-RhFcRn IV, and effects on total IgG, as well as DSA IgG, were measured, in addition to IgM and protective immunity. Subsequently, 60mg/kg IV was given in the setting of kidney transplantation from skin graft donors. Kidney transplant recipients received RhATG, and tacrolimus, MMF, and steroid for maintenance immunosuppression. Results Circulating total IgG was reduced from a baseline 100% on D0 to 32.0% (mean, SD ± 10.6) on d4 post infusion (p<0.05), while using a DSA assay. T-cell flow cross match (TFXM) was reduced to 40.6±12.5% of baseline, and B-cell FXCM to 52.2±19.3%. Circulating total IgM and DSA IgM were unaffected by treatment. Pathogen-specific antibodies (anti-gB and anti-tetanus toxin IgG) were significantly reduced for 14d post infusion. Post-transplant, circulating IgG responded to anti-FcRn mAb treatment, but DSA increased rapidly. Conclusion Targeting the FcRn-mediated recycling of IgG is an effective means of lowering circulating donor-specific IgG in the sensitized recipient, although in the setting of organ transplantation mechanisms of rapid antibody rise post-transplant remains unaffected.
Collapse
Affiliation(s)
- Miriam Manook
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Walter J Flores
- Massbiologics of the University of Massachusetts Medical School, Boston, MA, United States
| | - Robin Schmitz
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Zachary Fitch
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Janghoon Yoon
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Yeeun Bae
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Brian Shaw
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Allan Kirk
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Melissa Harnois
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States
| | - Sallie Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, United States
| | - Alton B Farris
- Department of Pathology, Emory School of Medicine, Atlanta, GA, United States
| | - Diogo M Magnani
- Massbiologics of the University of Massachusetts Medical School, Boston, MA, United States
| | - Jean Kwun
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Stuart Knechtle
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
195
|
Correlation of Donor-Derived Cell-free DNA with Histology and Molecular Diagnoses of Kidney Transplant Biopsies. Transplantation 2021; 106:1061-1070. [PMID: 34075006 DOI: 10.1097/tp.0000000000003838] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Circulating donor-derived cell free DNA (cfDNA), a minimally invasive diagnostic tool for kidney transplant rejection, was validated using traditional histology. The Molecular Microscope (MMDx) tissue gene expression platform may provide increased precision to traditional histology. METHODS In this single-center prospective study of 208 biopsies (median=5.8 months) post-transplant, we report on the calibration of cfDNA with simultaneous biopsy assessments using MMDx and histology by Area under the curve (AUC) analyses for optimal criterion, as well as for, previously published cfDNA cut-offs ≤0.21% to 'rule-out' rejection and ≥1% to 'rule-in' rejection. RESULTS There were significant discrepancies between histology and MMDx, with MMDx identifying more antibody-mediated rejection (65; 31%) than histology (43; 21%); the opposite was true for T-cell mediated rejection [TCMR; histology: 27 (13%) vs MMDx: 13 (6%)]. Most of the TCMR discrepancies were seen for histologic borderline/1A TCMR. AUC Curves for cfDNA and prediction of rejection were slightly better with MMDx (AUC=0.80; 95%CI: 0.74-0.86) vs. histology (AUC=0.75; 95%CI: 0.69-0.81). A cfDNA≤0.21% had similar sensitivity (~91%) to 'rule-out' rejection by histology and MMDx. Specificity was slightly higher with MMDx (92%) compared with histology (85%) to 'rule-in' rejection using cfDNA criterion≥1%. Strong positive quantitative correlations were observed between cfDNA scores and molecular acute kidney injury (AKI) for both 'rejection' and 'nonrejection' biopsies. CONCLUSIONS Molecular diagnostics using tissue gene expression and blood-based donor-derived cell-free DNA may add precision to some cases of traditional histology. The positive correlation of cfDNA with molecular AKI suggests a dose-dependent association with tissue injury irrespective of rejection characteristics.
Collapse
|
196
|
Halverson LP, Hachem RR. Antibody-Mediated Rejection and Lung Transplantation. Semin Respir Crit Care Med 2021; 42:428-435. [PMID: 34030204 DOI: 10.1055/s-0041-1728796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibody-mediated rejection (AMR) is now a widely recognized form of lung allograft rejection, with mounting evidence for AMR as an important risk factor for the development of chronic lung allograft dysfunction and markedly decreased long-term survival. Despite the recent development of the consensus diagnostic criteria, it remains a challenging diagnosis of exclusion. Furthermore, even after diagnosis, treatment directed at pulmonary AMR has been nearly exclusively derived from practices with other solid-organ transplants and other areas of medicine, such that there is a significant lack of data regarding the efficacy for these in pulmonary AMR. Lastly, outcomes after AMR remain quite poor despite aggressive treatment. In this review, we revisit the history of AMR in lung transplantation, describe our current understanding of its pathophysiology, discuss the use and limitations of the consensus diagnostic criteria, review current treatment strategies, and summarize long-term outcomes. We conclude with a synopsis of our most pressing gaps in knowledge, introduce recommendations for future directions, and highlight promising areas of active research.
Collapse
Affiliation(s)
- Laura P Halverson
- Division of Pulmonary and Critical Care, Washington University School of Medicine, Saint Louis, Missouri
| | - Ramsey R Hachem
- Division of Pulmonary and Critical Care, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
197
|
Amubieya O, Ramsey A, DerHovanessian A, Fishbein GA, Lynch JP, Belperio JA, Weigt SS. Chronic Lung Allograft Dysfunction: Evolving Concepts and Therapies. Semin Respir Crit Care Med 2021; 42:392-410. [PMID: 34030202 DOI: 10.1055/s-0041-1729175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The primary factor that limits long-term survival after lung transplantation is chronic lung allograft dysfunction (CLAD). CLAD also impairs quality of life and increases the costs of medical care. Our understanding of CLAD continues to evolve. Consensus definitions of CLAD and the major CLAD phenotypes were recently updated and clarified, but it remains to be seen whether the current definitions will lead to advances in management or impact care. Understanding the potential differences in pathogenesis for each CLAD phenotype may lead to novel therapeutic strategies, including precision medicine. Recognition of CLAD risk factors may lead to earlier interventions to mitigate risk, or to avoid risk factors all together, to prevent the development of CLAD. Unfortunately, currently available therapies for CLAD are usually not effective. However, novel therapeutics aimed at both prevention and treatment are currently under investigation. We provide an overview of the updates to CLAD-related terminology, clinical phenotypes and their diagnosis, natural history, pathogenesis, and potential strategies to treat and prevent CLAD.
Collapse
Affiliation(s)
- Olawale Amubieya
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Allison Ramsey
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ariss DerHovanessian
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gregory A Fishbein
- Department of Pathology, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Joseph P Lynch
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - John A Belperio
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - S Samuel Weigt
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Internal Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
198
|
Mrevlje M, Oblak M, Mlinšek G, Lindič J, Jadranka-Buturović-Ponikvar, Arnol M. First and second morning spot urine protein measurements for the assessment of proteinuria: a diagnostic accuracy study in kidney transplant recipients. BMC Nephrol 2021; 22:192. [PMID: 34022831 PMCID: PMC8141254 DOI: 10.1186/s12882-021-02406-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Quantification of proteinuria in kidney transplant recipients is important for diagnostic and prognostic purposes. Apart from correlation tests, there have been few evaluations of spot urine protein measurements in kidney transplantation. METHODS In this cross-sectional study involving 151 transplanted patients, we investigated measures of agreement (bias and accuracy) between the estimated protein excretion rate (ePER), determined from the protein-to-creatinine ratio in the first and second morning urine, and 24-h proteinuria and studied their performance at different levels of proteinuria. Measures of agreement were reanalyzed in relation to allograft histology in 76 patients with kidney biopsies performed for cause before enrolment in the study. RESULTS For ePER in the first morning urine, percent bias ranged from 1 to 28% and accuracy (within 30% of 24-h collection) ranged from 56 to 73%. For the second morning urine, percent bias ranged from 2 to 11%, and accuracy ranged from 71 to 78%. The accuracy of ePER (within 30%) in first and second morning urine progressively increased from 56 and 71% for low-grade proteinuria (150-299 mg/day) to 60 and 74% for moderate proteinuria (300-999 mg/day), and to 73 and 78% for high-grade proteinuria (≥1000 mg/day). Measures of agreement were similar across histologic phenotypes of allograft injury. CONCLUSIONS The ability of ePER to accurately predict 24-h proteinuria in kidney transplant recipients is modest. However, accuracy improves with an increase in proteinuria. Given the similar accuracy of ePER measurements in first and second morning urine, second morning urine can be used to monitor protein excretion.
Collapse
Affiliation(s)
- Maja Mrevlje
- Department of Nephrology, Centre for Kidney Transplantation, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia
- Department of internal medicine, General Hospital Izola, Izola, Slovenia
| | - Manca Oblak
- Department of Nephrology, Centre for Kidney Transplantation, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia
| | - Gregor Mlinšek
- Department of Nephrology, Centre for Kidney Transplantation, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia
| | - Jelka Lindič
- Department of Nephrology, Centre for Kidney Transplantation, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia
| | - Jadranka-Buturović-Ponikvar
- Department of Nephrology, Centre for Kidney Transplantation, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Miha Arnol
- Department of Nephrology, Centre for Kidney Transplantation, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia.
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
199
|
An Integrated Transcriptomic Approach to Identify Molecular Markers of Calcineurin Inhibitor Nephrotoxicity in Pediatric Kidney Transplant Recipients. Int J Mol Sci 2021; 22:ijms22115414. [PMID: 34063776 PMCID: PMC8196602 DOI: 10.3390/ijms22115414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 01/29/2023] Open
Abstract
Calcineurin inhibitors are highly efficacious immunosuppressive agents used in pediatric kidney transplantation. However, calcineurin inhibitor nephrotoxicity (CNIT) has been associated with the development of chronic renal allograft dysfunction and decreased graft survival. This study evaluated 37 formalin-fixed paraffin-embedded biopsies from pediatric kidney transplant recipients using gene expression profiling. Normal allograft samples (n = 12) served as negative controls and were compared to biopsies exhibiting CNIT (n = 11). The remaining samples served as positive controls to validate CNIT marker specificity and were characterized by other common causes of graft failure such as acute rejection (n = 7) and interstitial fibrosis/tubular atrophy (n = 7). MiRNA profiles served as the platform for data integration. Oxidative phosphorylation and mitochondrial dysfunction were the top molecular pathways associated with overexpressed genes in CNIT samples. Decreased ATP synthesis was identified as a significant biological function in CNIT, while key toxicology pathways included NRF2-mediated oxidative stress response and increased permeability transition of mitochondria. An integrative analysis demonstrated a panel of 13 significant miRNAs and their 33 CNIT-specific gene targets involved with mitochondrial activity and function. We also identified a candidate panel of miRNAs/genes, which may serve as future molecular markers for CNIT diagnosis as well as potential therapeutic targets.
Collapse
|
200
|
The role of non-HLA antibodies in solid organ transplantation: a complex deliberation. Curr Opin Organ Transplant 2021; 25:536-542. [PMID: 33044346 DOI: 10.1097/mot.0000000000000811] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW There is tremendous interest in understanding when, if, and how non-HLA antibodies contribute to allograft injury. Numerous non-HLA target antigens have been identified and sensitization to these targets have been associated with delayed allograft function, rejection, and allograft failure. This review focuses on the clinical utility of HLA antibody testing, highlighting the strengths and limitations of current clinical studies, and the need for defining characteristics to inform non-HLA antibody pathogenicity. RECENT FINDINGS Clinical studies continue to show associations between non-HLA antibodies and rejection and reduced allograft survival across multiple transplanted organ types. The worst clinical outcomes continue to be observed among recipients testing positive for both non-HLA and donor-specific HLA antibodies. Mechanistic insights from both animal and clinical studies support a model in which tissue injury accompanied by an inflammatory environment influence non-HLA antibody formation and pathogenicity. SUMMARY Immune triggers that lead to non-HLA antibody formation and pathogenicity are complex and poorly understood. The ability of non-HLA antibodies to mediate allograft injury may depend upon their affinity and strength (titer), target specificity, density of the target antigen, and synergy with donor-specific HLA antibodies.
Collapse
|